US20230364207A1 - Mutant of immunoglobulin degrading enzyme idee - Google Patents
Mutant of immunoglobulin degrading enzyme idee Download PDFInfo
- Publication number
- US20230364207A1 US20230364207A1 US18/001,876 US202118001876A US2023364207A1 US 20230364207 A1 US20230364207 A1 US 20230364207A1 US 202118001876 A US202118001876 A US 202118001876A US 2023364207 A1 US2023364207 A1 US 2023364207A1
- Authority
- US
- United States
- Prior art keywords
- mutant
- last
- idee
- immunoglobulin
- degrading enzyme
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
- 102000004190 Enzymes Human genes 0.000 title claims abstract description 83
- 108090000790 Enzymes Proteins 0.000 title claims abstract description 83
- 108060003951 Immunoglobulin Proteins 0.000 title abstract description 14
- 102000018358 immunoglobulin Human genes 0.000 title abstract description 14
- 230000000593 degrading effect Effects 0.000 title abstract 5
- 230000000694 effects Effects 0.000 claims abstract description 64
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 claims abstract description 33
- 108090000623 proteins and genes Proteins 0.000 claims description 73
- 235000001014 amino acid Nutrition 0.000 claims description 62
- 150000001413 amino acids Chemical group 0.000 claims description 58
- 229940024606 amino acid Drugs 0.000 claims description 57
- 102000004169 proteins and genes Human genes 0.000 claims description 47
- 235000018102 proteins Nutrition 0.000 claims description 45
- 239000003814 drug Substances 0.000 claims description 44
- 229940079593 drug Drugs 0.000 claims description 39
- 239000000203 mixture Substances 0.000 claims description 24
- 241000700605 Viruses Species 0.000 claims description 23
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 claims description 22
- 210000004027 cell Anatomy 0.000 claims description 19
- 238000012217 deletion Methods 0.000 claims description 19
- 230000037430 deletion Effects 0.000 claims description 19
- 230000035772 mutation Effects 0.000 claims description 19
- 239000013603 viral vector Substances 0.000 claims description 19
- 210000004369 blood Anatomy 0.000 claims description 17
- 239000008280 blood Substances 0.000 claims description 17
- 235000006109 methionine Nutrition 0.000 claims description 17
- 229930182817 methionine Natural products 0.000 claims description 17
- 229960004452 methionine Drugs 0.000 claims description 17
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 claims description 16
- 235000014304 histidine Nutrition 0.000 claims description 16
- 229960002885 histidine Drugs 0.000 claims description 16
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 claims description 16
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 claims description 15
- 235000004279 alanine Nutrition 0.000 claims description 15
- 239000004475 Arginine Substances 0.000 claims description 14
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 claims description 14
- KDXKERNSBIXSRK-YFKPBYRVSA-N L-lysine Chemical compound NCCCC[C@H](N)C(O)=O KDXKERNSBIXSRK-YFKPBYRVSA-N 0.000 claims description 14
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 claims description 14
- 239000004472 Lysine Substances 0.000 claims description 14
- AYFVYJQAPQTCCC-UHFFFAOYSA-N Threonine Natural products CC(O)C(N)C(O)=O AYFVYJQAPQTCCC-UHFFFAOYSA-N 0.000 claims description 14
- 239000004473 Threonine Substances 0.000 claims description 14
- 229960003121 arginine Drugs 0.000 claims description 14
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 claims description 14
- 235000009697 arginine Nutrition 0.000 claims description 14
- 235000013922 glutamic acid Nutrition 0.000 claims description 14
- 229960002989 glutamic acid Drugs 0.000 claims description 14
- 239000004220 glutamic acid Substances 0.000 claims description 14
- 235000018977 lysine Nutrition 0.000 claims description 14
- 229960003646 lysine Drugs 0.000 claims description 14
- 229960002898 threonine Drugs 0.000 claims description 14
- 235000008521 threonine Nutrition 0.000 claims description 14
- MTCFGRXMJLQNBG-REOHCLBHSA-N (2S)-2-Amino-3-hydroxypropansäure Chemical compound OC[C@H](N)C(O)=O MTCFGRXMJLQNBG-REOHCLBHSA-N 0.000 claims description 13
- DCXYFEDJOCDNAF-UHFFFAOYSA-N Asparagine Natural products OC(=O)C(N)CC(N)=O DCXYFEDJOCDNAF-UHFFFAOYSA-N 0.000 claims description 13
- 102100026120 IgG receptor FcRn large subunit p51 Human genes 0.000 claims description 13
- 101710177940 IgG receptor FcRn large subunit p51 Proteins 0.000 claims description 13
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 claims description 13
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 claims description 13
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 claims description 13
- QIVBCDIJIAJPQS-VIFPVBQESA-N L-tryptophane Chemical compound C1=CC=C2C(C[C@H](N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-VIFPVBQESA-N 0.000 claims description 13
- KZSNJWFQEVHDMF-BYPYZUCNSA-N L-valine Chemical compound CC(C)[C@H](N)C(O)=O KZSNJWFQEVHDMF-BYPYZUCNSA-N 0.000 claims description 13
- ROHFNLRQFUQHCH-UHFFFAOYSA-N Leucine Natural products CC(C)CC(N)C(O)=O ROHFNLRQFUQHCH-UHFFFAOYSA-N 0.000 claims description 13
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 claims description 13
- QIVBCDIJIAJPQS-UHFFFAOYSA-N Tryptophan Natural products C1=CC=C2C(CC(N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-UHFFFAOYSA-N 0.000 claims description 13
- KZSNJWFQEVHDMF-UHFFFAOYSA-N Valine Natural products CC(C)C(N)C(O)=O KZSNJWFQEVHDMF-UHFFFAOYSA-N 0.000 claims description 13
- 229960001230 asparagine Drugs 0.000 claims description 13
- 235000009582 asparagine Nutrition 0.000 claims description 13
- 229960005261 aspartic acid Drugs 0.000 claims description 13
- 235000003704 aspartic acid Nutrition 0.000 claims description 13
- OQFSQFPPLPISGP-UHFFFAOYSA-N beta-carboxyaspartic acid Natural products OC(=O)C(N)C(C(O)=O)C(O)=O OQFSQFPPLPISGP-UHFFFAOYSA-N 0.000 claims description 13
- 235000014705 isoleucine Nutrition 0.000 claims description 13
- 229960000310 isoleucine Drugs 0.000 claims description 13
- AGPKZVBTJJNPAG-UHFFFAOYSA-N isoleucine Natural products CCC(C)C(N)C(O)=O AGPKZVBTJJNPAG-UHFFFAOYSA-N 0.000 claims description 13
- 235000005772 leucine Nutrition 0.000 claims description 13
- 229960003136 leucine Drugs 0.000 claims description 13
- 229960001153 serine Drugs 0.000 claims description 13
- 235000004400 serine Nutrition 0.000 claims description 13
- 229960004799 tryptophan Drugs 0.000 claims description 13
- 229960004441 tyrosine Drugs 0.000 claims description 13
- 235000002374 tyrosine Nutrition 0.000 claims description 13
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 claims description 13
- 229960004295 valine Drugs 0.000 claims description 13
- 239000004474 valine Substances 0.000 claims description 13
- 235000014393 valine Nutrition 0.000 claims description 13
- XUJNEKJLAYXESH-REOHCLBHSA-N L-Cysteine Chemical compound SC[C@H](N)C(O)=O XUJNEKJLAYXESH-REOHCLBHSA-N 0.000 claims description 12
- ONIBWKKTOPOVIA-BYPYZUCNSA-N L-Proline Chemical compound OC(=O)[C@@H]1CCCN1 ONIBWKKTOPOVIA-BYPYZUCNSA-N 0.000 claims description 12
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-N 0.000 claims description 12
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 claims description 12
- AGPKZVBTJJNPAG-WHFBIAKZSA-N L-isoleucine Chemical compound CC[C@H](C)[C@H](N)C(O)=O AGPKZVBTJJNPAG-WHFBIAKZSA-N 0.000 claims description 12
- COLNVLDHVKWLRT-QMMMGPOBSA-N L-phenylalanine Chemical compound OC(=O)[C@@H](N)CC1=CC=CC=C1 COLNVLDHVKWLRT-QMMMGPOBSA-N 0.000 claims description 12
- ONIBWKKTOPOVIA-UHFFFAOYSA-N Proline Natural products OC(=O)C1CCCN1 ONIBWKKTOPOVIA-UHFFFAOYSA-N 0.000 claims description 12
- 229960002433 cysteine Drugs 0.000 claims description 12
- 235000018417 cysteine Nutrition 0.000 claims description 12
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 claims description 12
- 235000004554 glutamine Nutrition 0.000 claims description 12
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 claims description 12
- 229960002743 glutamine Drugs 0.000 claims description 12
- 235000008729 phenylalanine Nutrition 0.000 claims description 12
- 229960005190 phenylalanine Drugs 0.000 claims description 12
- COLNVLDHVKWLRT-UHFFFAOYSA-N phenylalanine Natural products OC(=O)C(N)CC1=CC=CC=C1 COLNVLDHVKWLRT-UHFFFAOYSA-N 0.000 claims description 12
- 229960002429 proline Drugs 0.000 claims description 12
- 235000013930 proline Nutrition 0.000 claims description 12
- 238000006467 substitution reaction Methods 0.000 claims description 12
- 239000004471 Glycine Substances 0.000 claims description 11
- ODKSFYDXXFIFQN-BYPYZUCNSA-P L-argininium(2+) Chemical compound NC(=[NH2+])NCCC[C@H]([NH3+])C(O)=O ODKSFYDXXFIFQN-BYPYZUCNSA-P 0.000 claims description 11
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 claims description 11
- 229960002449 glycine Drugs 0.000 claims description 11
- 102000018071 Immunoglobulin Fc Fragments Human genes 0.000 claims description 10
- 108010091135 Immunoglobulin Fc Fragments Proteins 0.000 claims description 10
- 238000001415 gene therapy Methods 0.000 claims description 10
- 239000002773 nucleotide Substances 0.000 claims description 10
- 125000003729 nucleotide group Chemical group 0.000 claims description 10
- 244000309459 oncolytic virus Species 0.000 claims description 10
- 229960003767 alanine Drugs 0.000 claims description 9
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 claims description 8
- AYFVYJQAPQTCCC-GBXIJSLDSA-N L-threonine Chemical compound C[C@@H](O)[C@H](N)C(O)=O AYFVYJQAPQTCCC-GBXIJSLDSA-N 0.000 claims description 8
- 125000003295 alanine group Chemical group N[C@@H](C)C(=O)* 0.000 claims description 7
- 239000003795 chemical substances by application Substances 0.000 claims description 7
- 239000003937 drug carrier Substances 0.000 claims description 7
- 239000013604 expression vector Substances 0.000 claims description 7
- 102000004127 Cytokines Human genes 0.000 claims description 6
- 108090000695 Cytokines Proteins 0.000 claims description 6
- 125000000341 threoninyl group Chemical group [H]OC([H])(C([H])([H])[H])C([H])(N([H])[H])C(*)=O 0.000 claims description 6
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 5
- 102000037982 Immune checkpoint proteins Human genes 0.000 claims description 4
- 108091008036 Immune checkpoint proteins Proteins 0.000 claims description 4
- 229940023147 viral vector vaccine Drugs 0.000 claims description 4
- 102000018697 Membrane Proteins Human genes 0.000 claims description 3
- 108010052285 Membrane Proteins Proteins 0.000 claims description 3
- 125000000637 arginyl group Chemical group N[C@@H](CCCNC(N)=N)C(=O)* 0.000 claims description 3
- 125000000291 glutamic acid group Chemical group N[C@@H](CCC(O)=O)C(=O)* 0.000 claims description 3
- 229940088597 hormone Drugs 0.000 claims description 3
- 239000005556 hormone Substances 0.000 claims description 3
- 230000003834 intracellular effect Effects 0.000 claims description 3
- 241000588724 Escherichia coli Species 0.000 claims description 2
- 210000005253 yeast cell Anatomy 0.000 claims description 2
- 125000003275 alpha amino acid group Chemical group 0.000 abstract description 24
- 238000003776 cleavage reaction Methods 0.000 description 83
- 230000007017 scission Effects 0.000 description 74
- 229940088598 enzyme Drugs 0.000 description 57
- 239000000047 product Substances 0.000 description 41
- 210000002966 serum Anatomy 0.000 description 36
- 229940027941 immunoglobulin g Drugs 0.000 description 31
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 30
- 230000006862 enzymatic digestion Effects 0.000 description 22
- 238000000034 method Methods 0.000 description 20
- 108090000765 processed proteins & peptides Proteins 0.000 description 19
- 239000000758 substrate Substances 0.000 description 19
- 241000699670 Mus sp. Species 0.000 description 18
- 229920001184 polypeptide Polymers 0.000 description 18
- 102000004196 processed proteins & peptides Human genes 0.000 description 18
- 241000699666 Mus <mouse, genus> Species 0.000 description 17
- 238000006243 chemical reaction Methods 0.000 description 17
- 238000001962 electrophoresis Methods 0.000 description 16
- 229960000575 trastuzumab Drugs 0.000 description 16
- 238000001502 gel electrophoresis Methods 0.000 description 14
- 239000012160 loading buffer Substances 0.000 description 13
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 13
- 239000006228 supernatant Substances 0.000 description 12
- 210000002381 plasma Anatomy 0.000 description 11
- 201000010099 disease Diseases 0.000 description 9
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 9
- 102000040430 polynucleotide Human genes 0.000 description 9
- 108091033319 polynucleotide Proteins 0.000 description 9
- 239000002157 polynucleotide Substances 0.000 description 9
- -1 CD79 Proteins 0.000 description 8
- 238000002965 ELISA Methods 0.000 description 8
- 102100039620 Granulocyte-macrophage colony-stimulating factor Human genes 0.000 description 8
- 241000282412 Homo Species 0.000 description 8
- 108010021466 Mutant Proteins Proteins 0.000 description 8
- 102000008300 Mutant Proteins Human genes 0.000 description 8
- 241000283973 Oryctolagus cuniculus Species 0.000 description 8
- 239000013612 plasmid Substances 0.000 description 8
- 108010076504 Protein Sorting Signals Proteins 0.000 description 7
- 238000001514 detection method Methods 0.000 description 7
- 241000894007 species Species 0.000 description 7
- 241000700584 Simplexvirus Species 0.000 description 6
- 239000002246 antineoplastic agent Substances 0.000 description 6
- 210000004899 c-terminal region Anatomy 0.000 description 6
- 229940044683 chemotherapy drug Drugs 0.000 description 6
- UQLDLKMNUJERMK-UHFFFAOYSA-L di(octadecanoyloxy)lead Chemical compound [Pb+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O UQLDLKMNUJERMK-UHFFFAOYSA-L 0.000 description 6
- 229940126533 immune checkpoint blocker Drugs 0.000 description 6
- 239000007788 liquid Substances 0.000 description 6
- 150000007523 nucleic acids Chemical class 0.000 description 6
- 239000000243 solution Substances 0.000 description 6
- 229940045513 CTLA4 antagonist Drugs 0.000 description 5
- 241000672609 Escherichia coli BL21 Species 0.000 description 5
- 108010032606 Fragile X Mental Retardation Protein Proteins 0.000 description 5
- 241001465754 Metazoa Species 0.000 description 5
- 206010028980 Neoplasm Diseases 0.000 description 5
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 5
- 102100023532 Synaptic functional regulator FMR1 Human genes 0.000 description 5
- 229940126547 T-cell immunoglobulin mucin-3 Drugs 0.000 description 5
- AVKUERGKIZMTKX-NJBDSQKTSA-N ampicillin Chemical compound C1([C@@H](N)C(=O)N[C@H]2[C@H]3SC([C@@H](N3C2=O)C(O)=O)(C)C)=CC=CC=C1 AVKUERGKIZMTKX-NJBDSQKTSA-N 0.000 description 5
- 229960000723 ampicillin Drugs 0.000 description 5
- 230000002238 attenuated effect Effects 0.000 description 5
- 102200129340 c.176C>T Human genes 0.000 description 5
- 229940072221 immunoglobulins Drugs 0.000 description 5
- 238000012986 modification Methods 0.000 description 5
- 230000004048 modification Effects 0.000 description 5
- 102000039446 nucleic acids Human genes 0.000 description 5
- 108020004707 nucleic acids Proteins 0.000 description 5
- 238000000746 purification Methods 0.000 description 5
- 102200095430 rs28940294 Human genes 0.000 description 5
- 102220083823 rs863224587 Human genes 0.000 description 5
- 230000003248 secreting effect Effects 0.000 description 5
- 229940124597 therapeutic agent Drugs 0.000 description 5
- 108020004705 Codon Proteins 0.000 description 4
- 102220542356 Endogenous retrovirus group K member 113 Pro protein_T24A_mutation Human genes 0.000 description 4
- 102000014150 Interferons Human genes 0.000 description 4
- 108010050904 Interferons Proteins 0.000 description 4
- 241000700159 Rattus Species 0.000 description 4
- 241000702263 Reovirus sp. Species 0.000 description 4
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 4
- 241000700618 Vaccinia virus Species 0.000 description 4
- 239000000427 antigen Substances 0.000 description 4
- 108091007433 antigens Proteins 0.000 description 4
- 102000036639 antigens Human genes 0.000 description 4
- 229960003444 immunosuppressant agent Drugs 0.000 description 4
- 239000003018 immunosuppressive agent Substances 0.000 description 4
- 238000000338 in vitro Methods 0.000 description 4
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 description 4
- JDNTWHVOXJZDSN-UHFFFAOYSA-N iodoacetic acid Chemical compound OC(=O)CI JDNTWHVOXJZDSN-UHFFFAOYSA-N 0.000 description 4
- 125000001360 methionine group Chemical group N[C@@H](CCSC)C(=O)* 0.000 description 4
- 229940126619 mouse monoclonal antibody Drugs 0.000 description 4
- 229920000642 polymer Polymers 0.000 description 4
- 102220053185 rs727504747 Human genes 0.000 description 4
- 102000003390 tumor necrosis factor Human genes 0.000 description 4
- 241000701161 unidentified adenovirus Species 0.000 description 4
- UEJJHQNACJXSKW-UHFFFAOYSA-N 2-(2,6-dioxopiperidin-3-yl)-1H-isoindole-1,3(2H)-dione Chemical compound O=C1C2=CC=CC=C2C(=O)N1C1CCC(=O)NC1=O UEJJHQNACJXSKW-UHFFFAOYSA-N 0.000 description 3
- 241000711404 Avian avulavirus 1 Species 0.000 description 3
- 108010071942 Colony-Stimulating Factors Proteins 0.000 description 3
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 3
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 3
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- 239000002147 L01XE04 - Sunitinib Substances 0.000 description 3
- 102000038030 PI3Ks Human genes 0.000 description 3
- 108091007960 PI3Ks Proteins 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- 229940079156 Proteasome inhibitor Drugs 0.000 description 3
- 201000003176 Severe Acute Respiratory Syndrome Diseases 0.000 description 3
- 210000001744 T-lymphocyte Anatomy 0.000 description 3
- 102000013530 TOR Serine-Threonine Kinases Human genes 0.000 description 3
- 108010065917 TOR Serine-Threonine Kinases Proteins 0.000 description 3
- BPEGJWRSRHCHSN-UHFFFAOYSA-N Temozolomide Chemical compound O=C1N(C)N=NC2=C(C(N)=O)N=CN21 BPEGJWRSRHCHSN-UHFFFAOYSA-N 0.000 description 3
- 238000010306 acid treatment Methods 0.000 description 3
- 125000000539 amino acid group Chemical group 0.000 description 3
- 238000003556 assay Methods 0.000 description 3
- 229960001467 bortezomib Drugs 0.000 description 3
- GXJABQQUPOEUTA-RDJZCZTQSA-N bortezomib Chemical compound C([C@@H](C(=O)N[C@@H](CC(C)C)B(O)O)NC(=O)C=1N=CC=NC=1)C1=CC=CC=C1 GXJABQQUPOEUTA-RDJZCZTQSA-N 0.000 description 3
- 201000011510 cancer Diseases 0.000 description 3
- 230000022131 cell cycle Effects 0.000 description 3
- 229960004397 cyclophosphamide Drugs 0.000 description 3
- 229940127089 cytotoxic agent Drugs 0.000 description 3
- 239000002254 cytotoxic agent Substances 0.000 description 3
- 239000003085 diluting agent Substances 0.000 description 3
- 239000000539 dimer Substances 0.000 description 3
- 241001493065 dsRNA viruses Species 0.000 description 3
- 230000001973 epigenetic effect Effects 0.000 description 3
- SDUQYLNIPVEERB-QPPQHZFASA-N gemcitabine Chemical compound O=C1N=C(N)C=CN1[C@H]1C(F)(F)[C@H](O)[C@@H](CO)O1 SDUQYLNIPVEERB-QPPQHZFASA-N 0.000 description 3
- 229960005277 gemcitabine Drugs 0.000 description 3
- 229940121372 histone deacetylase inhibitor Drugs 0.000 description 3
- 239000003276 histone deacetylase inhibitor Substances 0.000 description 3
- 230000001861 immunosuppressant effect Effects 0.000 description 3
- 238000001727 in vivo Methods 0.000 description 3
- 239000003112 inhibitor Substances 0.000 description 3
- 229940079322 interferon Drugs 0.000 description 3
- 238000004519 manufacturing process Methods 0.000 description 3
- 229960001156 mitoxantrone Drugs 0.000 description 3
- KKZJGLLVHKMTCM-UHFFFAOYSA-N mitoxantrone Chemical compound O=C1C2=C(O)C=CC(O)=C2C(=O)C2=C1C(NCCNCCO)=CC=C2NCCNCCO KKZJGLLVHKMTCM-UHFFFAOYSA-N 0.000 description 3
- 239000000178 monomer Substances 0.000 description 3
- 230000003287 optical effect Effects 0.000 description 3
- 244000052769 pathogen Species 0.000 description 3
- 230000001717 pathogenic effect Effects 0.000 description 3
- 239000008194 pharmaceutical composition Substances 0.000 description 3
- 229960000688 pomalidomide Drugs 0.000 description 3
- UVSMNLNDYGZFPF-UHFFFAOYSA-N pomalidomide Chemical compound O=C1C=2C(N)=CC=CC=2C(=O)N1C1CCC(=O)NC1=O UVSMNLNDYGZFPF-UHFFFAOYSA-N 0.000 description 3
- 239000003207 proteasome inhibitor Substances 0.000 description 3
- 238000012216 screening Methods 0.000 description 3
- 238000012772 sequence design Methods 0.000 description 3
- 230000019491 signal transduction Effects 0.000 description 3
- 239000011780 sodium chloride Substances 0.000 description 3
- 239000000126 substance Substances 0.000 description 3
- WINHZLLDWRZWRT-ATVHPVEESA-N sunitinib Chemical compound CCN(CC)CCNC(=O)C1=C(C)NC(\C=C/2C3=CC(F)=CC=C3NC\2=O)=C1C WINHZLLDWRZWRT-ATVHPVEESA-N 0.000 description 3
- 229960001796 sunitinib Drugs 0.000 description 3
- 239000000725 suspension Substances 0.000 description 3
- 230000008685 targeting Effects 0.000 description 3
- 229960004964 temozolomide Drugs 0.000 description 3
- 238000012360 testing method Methods 0.000 description 3
- 229960003433 thalidomide Drugs 0.000 description 3
- 229940121358 tyrosine kinase inhibitor Drugs 0.000 description 3
- 239000005483 tyrosine kinase inhibitor Substances 0.000 description 3
- 150000004917 tyrosine kinase inhibitor derivatives Chemical class 0.000 description 3
- YBJHBAHKTGYVGT-ZKWXMUAHSA-N (+)-Biotin Chemical compound N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 YBJHBAHKTGYVGT-ZKWXMUAHSA-N 0.000 description 2
- 102100024378 AF4/FMR2 family member 2 Human genes 0.000 description 2
- 229920000936 Agarose Polymers 0.000 description 2
- 108700028369 Alleles Proteins 0.000 description 2
- 241000710929 Alphavirus Species 0.000 description 2
- 101710095342 Apolipoprotein B Proteins 0.000 description 2
- 102100040202 Apolipoprotein B-100 Human genes 0.000 description 2
- 108010029697 CD40 Ligand Proteins 0.000 description 2
- 102100032937 CD40 ligand Human genes 0.000 description 2
- 108010021064 CTLA-4 Antigen Proteins 0.000 description 2
- 241000282472 Canis lupus familiaris Species 0.000 description 2
- 241000282693 Cercopithecidae Species 0.000 description 2
- 102000019034 Chemokines Human genes 0.000 description 2
- 108010012236 Chemokines Proteins 0.000 description 2
- 102000012437 Copper-Transporting ATPases Human genes 0.000 description 2
- 108010022637 Copper-Transporting ATPases Proteins 0.000 description 2
- 241000709675 Coxsackievirus B3 Species 0.000 description 2
- 102100039498 Cytotoxic T-lymphocyte protein 4 Human genes 0.000 description 2
- 102000018651 Epithelial Cell Adhesion Molecule Human genes 0.000 description 2
- 108010066687 Epithelial Cell Adhesion Molecule Proteins 0.000 description 2
- 241000283073 Equus caballus Species 0.000 description 2
- 102000004547 Glucosylceramidase Human genes 0.000 description 2
- 108010017544 Glucosylceramidase Proteins 0.000 description 2
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 description 2
- 102100034458 Hepatitis A virus cellular receptor 2 Human genes 0.000 description 2
- 101710083479 Hepatitis A virus cellular receptor 2 homolog Proteins 0.000 description 2
- 101001137987 Homo sapiens Lymphocyte activation gene 3 protein Proteins 0.000 description 2
- 101000701517 Homo sapiens Putative protein ATXN8OS Proteins 0.000 description 2
- 101001012157 Homo sapiens Receptor tyrosine-protein kinase erbB-2 Proteins 0.000 description 2
- 101000709472 Homo sapiens Sialic acid-binding Ig-like lectin 15 Proteins 0.000 description 2
- 101000831007 Homo sapiens T-cell immunoreceptor with Ig and ITIM domains Proteins 0.000 description 2
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 description 2
- 101000914484 Homo sapiens T-lymphocyte activation antigen CD80 Proteins 0.000 description 2
- 101000904724 Homo sapiens Transmembrane glycoprotein NMB Proteins 0.000 description 2
- 101000801234 Homo sapiens Tumor necrosis factor receptor superfamily member 18 Proteins 0.000 description 2
- 101000851370 Homo sapiens Tumor necrosis factor receptor superfamily member 9 Proteins 0.000 description 2
- 101000666896 Homo sapiens V-type immunoglobulin domain-containing suppressor of T-cell activation Proteins 0.000 description 2
- 241000725303 Human immunodeficiency virus Species 0.000 description 2
- 102000004877 Insulin Human genes 0.000 description 2
- 108090001061 Insulin Proteins 0.000 description 2
- 108010002352 Interleukin-1 Proteins 0.000 description 2
- 108010065805 Interleukin-12 Proteins 0.000 description 2
- 102000013462 Interleukin-12 Human genes 0.000 description 2
- 108010002350 Interleukin-2 Proteins 0.000 description 2
- 102000000588 Interleukin-2 Human genes 0.000 description 2
- 102000017578 LAG3 Human genes 0.000 description 2
- 241000282567 Macaca fascicularis Species 0.000 description 2
- 241000711386 Mumps virus Species 0.000 description 2
- 108010052185 Myotonin-Protein Kinase Proteins 0.000 description 2
- 102100022437 Myotonin-protein kinase Human genes 0.000 description 2
- 108010025020 Nerve Growth Factor Proteins 0.000 description 2
- 108091028043 Nucleic acid sequence Proteins 0.000 description 2
- 241000283977 Oryctolagus Species 0.000 description 2
- 108091093037 Peptide nucleic acid Proteins 0.000 description 2
- 241000709664 Picornaviridae Species 0.000 description 2
- 101710089372 Programmed cell death protein 1 Proteins 0.000 description 2
- 102100040678 Programmed cell death protein 1 Human genes 0.000 description 2
- 102100030469 Putative protein ATXN8OS Human genes 0.000 description 2
- 102100030086 Receptor tyrosine-protein kinase erbB-2 Human genes 0.000 description 2
- 102100034361 Sialic acid-binding Ig-like lectin 15 Human genes 0.000 description 2
- 241000193996 Streptococcus pyogenes Species 0.000 description 2
- 241000282887 Suidae Species 0.000 description 2
- 102000008221 Superoxide Dismutase-1 Human genes 0.000 description 2
- 108010021188 Superoxide Dismutase-1 Proteins 0.000 description 2
- 108091008874 T cell receptors Proteins 0.000 description 2
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 2
- 102100024834 T-cell immunoreceptor with Ig and ITIM domains Human genes 0.000 description 2
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 description 2
- 102100027222 T-lymphocyte activation antigen CD80 Human genes 0.000 description 2
- 102000006601 Thymidine Kinase Human genes 0.000 description 2
- 108020004440 Thymidine kinase Proteins 0.000 description 2
- 102000009618 Transforming Growth Factors Human genes 0.000 description 2
- 108010009583 Transforming Growth Factors Proteins 0.000 description 2
- 102100023935 Transmembrane glycoprotein NMB Human genes 0.000 description 2
- 102100033728 Tumor necrosis factor receptor superfamily member 18 Human genes 0.000 description 2
- 102100022153 Tumor necrosis factor receptor superfamily member 4 Human genes 0.000 description 2
- 101710165473 Tumor necrosis factor receptor superfamily member 4 Proteins 0.000 description 2
- 102100036856 Tumor necrosis factor receptor superfamily member 9 Human genes 0.000 description 2
- 102100038282 V-type immunoglobulin domain-containing suppressor of T-cell activation Human genes 0.000 description 2
- 102000005789 Vascular Endothelial Growth Factors Human genes 0.000 description 2
- 108010019530 Vascular Endothelial Growth Factors Proteins 0.000 description 2
- 239000004480 active ingredient Substances 0.000 description 2
- 230000000692 anti-sense effect Effects 0.000 description 2
- 239000000969 carrier Substances 0.000 description 2
- 238000005119 centrifugation Methods 0.000 description 2
- 238000013461 design Methods 0.000 description 2
- 230000029087 digestion Effects 0.000 description 2
- 241001492478 dsDNA viruses, no RNA stage Species 0.000 description 2
- 230000002255 enzymatic effect Effects 0.000 description 2
- 238000002474 experimental method Methods 0.000 description 2
- IJJVMEJXYNJXOJ-UHFFFAOYSA-N fluquinconazole Chemical compound C=1C=C(Cl)C=C(Cl)C=1N1C(=O)C2=CC(F)=CC=C2N=C1N1C=NC=N1 IJJVMEJXYNJXOJ-UHFFFAOYSA-N 0.000 description 2
- 239000012634 fragment Substances 0.000 description 2
- 230000030279 gene silencing Effects 0.000 description 2
- 238000010438 heat treatment Methods 0.000 description 2
- 208000002672 hepatitis B Diseases 0.000 description 2
- 230000001506 immunosuppresive effect Effects 0.000 description 2
- 208000015181 infectious disease Diseases 0.000 description 2
- 230000002401 inhibitory effect Effects 0.000 description 2
- 238000002347 injection Methods 0.000 description 2
- 239000007924 injection Substances 0.000 description 2
- 229940125396 insulin Drugs 0.000 description 2
- 102000006495 integrins Human genes 0.000 description 2
- 108010044426 integrins Proteins 0.000 description 2
- 229940117681 interleukin-12 Drugs 0.000 description 2
- BPHPUYQFMNQIOC-NXRLNHOXSA-N isopropyl beta-D-thiogalactopyranoside Chemical compound CC(C)S[C@@H]1O[C@H](CO)[C@H](O)[C@H](O)[C@H]1O BPHPUYQFMNQIOC-NXRLNHOXSA-N 0.000 description 2
- 239000000463 material Substances 0.000 description 2
- 244000309715 mini pig Species 0.000 description 2
- 108010068617 neonatal Fc receptor Proteins 0.000 description 2
- 239000003921 oil Substances 0.000 description 2
- 235000019198 oils Nutrition 0.000 description 2
- 229960005547 pelareorep Drugs 0.000 description 2
- 238000002360 preparation method Methods 0.000 description 2
- 239000011535 reaction buffer Substances 0.000 description 2
- 241001147420 ssDNA viruses Species 0.000 description 2
- 241000114864 ssRNA viruses Species 0.000 description 2
- 241000712461 unidentified influenza virus Species 0.000 description 2
- 241001430294 unidentified retrovirus Species 0.000 description 2
- 239000013598 vector Substances 0.000 description 2
- NMWKYTGJWUAZPZ-WWHBDHEGSA-N (4S)-4-[[(4R,7S,10S,16S,19S,25S,28S,31R)-31-[[(2S)-2-[[(1R,6R,9S,12S,18S,21S,24S,27S,30S,33S,36S,39S,42R,47R,53S,56S,59S,62S,65S,68S,71S,76S,79S,85S)-47-[[(2S)-2-[[(2S)-4-amino-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-amino-3-methylbutanoyl]amino]-3-methylbutanoyl]amino]-3-hydroxypropanoyl]amino]-3-(1H-imidazol-4-yl)propanoyl]amino]-3-phenylpropanoyl]amino]-4-oxobutanoyl]amino]-3-carboxypropanoyl]amino]-18-(4-aminobutyl)-27,68-bis(3-amino-3-oxopropyl)-36,71,76-tribenzyl-39-(3-carbamimidamidopropyl)-24-(2-carboxyethyl)-21,56-bis(carboxymethyl)-65,85-bis[(1R)-1-hydroxyethyl]-59-(hydroxymethyl)-62,79-bis(1H-imidazol-4-ylmethyl)-9-methyl-33-(2-methylpropyl)-8,11,17,20,23,26,29,32,35,38,41,48,54,57,60,63,66,69,72,74,77,80,83,86-tetracosaoxo-30-propan-2-yl-3,4,44,45-tetrathia-7,10,16,19,22,25,28,31,34,37,40,49,55,58,61,64,67,70,73,75,78,81,84,87-tetracosazatetracyclo[40.31.14.012,16.049,53]heptaoctacontane-6-carbonyl]amino]-3-methylbutanoyl]amino]-7-(3-carbamimidamidopropyl)-25-(hydroxymethyl)-19-[(4-hydroxyphenyl)methyl]-28-(1H-imidazol-4-ylmethyl)-10-methyl-6,9,12,15,18,21,24,27,30-nonaoxo-16-propan-2-yl-1,2-dithia-5,8,11,14,17,20,23,26,29-nonazacyclodotriacontane-4-carbonyl]amino]-5-[[(2S)-1-[[(2S)-1-[[(2S)-3-carboxy-1-[[(2S)-1-[[(2S)-1-[[(1S)-1-carboxyethyl]amino]-4-methyl-1-oxopentan-2-yl]amino]-4-methyl-1-oxopentan-2-yl]amino]-1-oxopropan-2-yl]amino]-1-oxopropan-2-yl]amino]-3-(1H-imidazol-4-yl)-1-oxopropan-2-yl]amino]-5-oxopentanoic acid Chemical compound CC(C)C[C@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](C)NC(=O)[C@H](Cc1c[nH]cn1)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@@H]1CSSC[C@H](NC(=O)[C@@H](NC(=O)[C@@H]2CSSC[C@@H]3NC(=O)[C@H](Cc4ccccc4)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](NC(=O)[C@H](Cc4c[nH]cn4)NC(=O)[C@H](CO)NC(=O)[C@H](CC(O)=O)NC(=O)[C@@H]4CCCN4C(=O)[C@H](CSSC[C@H](NC(=O)[C@@H](NC(=O)CNC(=O)[C@H](Cc4c[nH]cn4)NC(=O)[C@H](Cc4ccccc4)NC3=O)[C@@H](C)O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](Cc3ccccc3)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCCCN)C(=O)N3CCC[C@H]3C(=O)N[C@@H](C)C(=O)N2)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](Cc2ccccc2)NC(=O)[C@H](Cc2c[nH]cn2)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@@H](N)C(C)C)C(C)C)[C@@H](C)O)C(C)C)C(=O)N[C@@H](Cc2c[nH]cn2)C(=O)N[C@@H](CO)C(=O)NCC(=O)N[C@@H](Cc2ccc(O)cc2)C(=O)N[C@@H](C(C)C)C(=O)NCC(=O)N[C@@H](C)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N1)C(=O)N[C@@H](C)C(O)=O NMWKYTGJWUAZPZ-WWHBDHEGSA-N 0.000 description 1
- SSOORFWOBGFTHL-OTEJMHTDSA-N (4S)-5-[[(2S)-1-[[(2S)-1-[[(2S)-1-[[(2S)-1-[[(2S)-1-[[(2S)-1-[[(2S)-1-[[(2S)-6-amino-1-[[(2S)-1-[[(2S)-1-[[(2S)-1-[[(2S)-1-[[2-[(2S)-2-[[(2S)-1-[[(2S)-1-[[(2S)-1-[[(2S)-1-[[(2S)-1-[[(2S)-1-[[(2S)-6-amino-1-[[(2S)-1-[[(2S)-1-[[(2S,3S)-1-[[(2S)-1-[[(2S)-1-[[(2S)-6-amino-1-[[(2S)-1-[[(2S)-1-[[(2S)-1-[[(2S)-1-[[(2S)-1-[[(2S)-5-amino-1-[[(2S)-1-[[(2S)-1-[[(2S)-6-amino-1-[[(2S)-6-amino-1-[[(2S)-1-[[(2S)-1-[[(2S)-5-amino-1-[[(2S)-5-carbamimidamido-1-[[(2S)-5-carbamimidamido-1-[[(1S)-4-carbamimidamido-1-carboxybutyl]amino]-1-oxopentan-2-yl]amino]-1-oxopentan-2-yl]amino]-1,5-dioxopentan-2-yl]amino]-5-carbamimidamido-1-oxopentan-2-yl]amino]-5-carbamimidamido-1-oxopentan-2-yl]amino]-1-oxohexan-2-yl]amino]-1-oxohexan-2-yl]amino]-5-carbamimidamido-1-oxopentan-2-yl]amino]-4-methyl-1-oxopentan-2-yl]amino]-1,5-dioxopentan-2-yl]amino]-4-methyl-1-oxopentan-2-yl]amino]-3-hydroxy-1-oxopropan-2-yl]amino]-3-hydroxy-1-oxopropan-2-yl]amino]-3-hydroxy-1-oxopropan-2-yl]amino]-1-oxopropan-2-yl]amino]-1-oxohexan-2-yl]amino]-3-hydroxy-1-oxopropan-2-yl]amino]-1-oxo-3-phenylpropan-2-yl]amino]-3-methyl-1-oxopentan-2-yl]amino]-3-methyl-1-oxobutan-2-yl]amino]-5-carbamimidamido-1-oxopentan-2-yl]amino]-1-oxohexan-2-yl]amino]-3-methyl-1-oxobutan-2-yl]amino]-5-carbamimidamido-1-oxopentan-2-yl]amino]-3-methyl-1-oxobutan-2-yl]amino]-4-methyl-1-oxopentan-2-yl]amino]-1-oxopropan-2-yl]amino]-5-carbamimidamido-1-oxopentan-2-yl]carbamoyl]pyrrolidin-1-yl]-2-oxoethyl]amino]-3-(1H-indol-3-yl)-1-oxopropan-2-yl]amino]-4-methyl-1-oxopentan-2-yl]amino]-1-oxo-3-phenylpropan-2-yl]amino]-5-carbamimidamido-1-oxopentan-2-yl]amino]-1-oxohexan-2-yl]amino]-3-methyl-1-oxobutan-2-yl]amino]-5-carbamimidamido-1-oxopentan-2-yl]amino]-4-methyl-1-oxopentan-2-yl]amino]-1-oxo-3-phenylpropan-2-yl]amino]-3-(1H-imidazol-4-yl)-1-oxopropan-2-yl]amino]-3-methyl-1-oxobutan-2-yl]amino]-4-methyl-1-oxopentan-2-yl]amino]-4-[[(2S)-2-[[(2S)-2-[[(2S)-2,6-diaminohexanoyl]amino]-3-methylbutanoyl]amino]propanoyl]amino]-5-oxopentanoic acid Chemical compound CC[C@H](C)[C@H](NC(=O)[C@@H](NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CCCCN)NC(=O)[C@@H](NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@H](C)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@@H]1CCCN1C(=O)CNC(=O)[C@H](Cc1c[nH]c2ccccc12)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](Cc1ccccc1)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CCCCN)NC(=O)[C@@H](NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](Cc1ccccc1)NC(=O)[C@H](Cc1c[nH]cn1)NC(=O)[C@@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](C)NC(=O)[C@@H](NC(=O)[C@@H](N)CCCCN)C(C)C)C(C)C)C(C)C)C(C)C)C(C)C)C(C)C)C(=O)N[C@@H](Cc1ccccc1)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](C)C(=O)N[C@@H](CO)C(=O)N[C@@H](CO)C(=O)N[C@@H](CO)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCCNC(N)=N)C(O)=O SSOORFWOBGFTHL-OTEJMHTDSA-N 0.000 description 1
- VGONTNSXDCQUGY-RRKCRQDMSA-N 2'-deoxyinosine Chemical group C1[C@H](O)[C@@H](CO)O[C@H]1N1C(N=CNC2=O)=C2N=C1 VGONTNSXDCQUGY-RRKCRQDMSA-N 0.000 description 1
- BGFTWECWAICPDG-UHFFFAOYSA-N 2-[bis(4-chlorophenyl)methyl]-4-n-[3-[bis(4-chlorophenyl)methyl]-4-(dimethylamino)phenyl]-1-n,1-n-dimethylbenzene-1,4-diamine Chemical compound C1=C(C(C=2C=CC(Cl)=CC=2)C=2C=CC(Cl)=CC=2)C(N(C)C)=CC=C1NC(C=1)=CC=C(N(C)C)C=1C(C=1C=CC(Cl)=CC=1)C1=CC=C(Cl)C=C1 BGFTWECWAICPDG-UHFFFAOYSA-N 0.000 description 1
- 108010046716 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide) Proteins 0.000 description 1
- 101150096316 5 gene Proteins 0.000 description 1
- 101710169336 5'-deoxyadenosine deaminase Proteins 0.000 description 1
- 101710184468 AF4/FMR2 family member 2 Proteins 0.000 description 1
- 101150091481 ATP7 gene Proteins 0.000 description 1
- 102100036664 Adenosine deaminase Human genes 0.000 description 1
- 102000002572 Alpha-Globulins Human genes 0.000 description 1
- 108010068307 Alpha-Globulins Proteins 0.000 description 1
- 208000024827 Alzheimer disease Diseases 0.000 description 1
- 102100032187 Androgen receptor Human genes 0.000 description 1
- 102000009840 Angiopoietins Human genes 0.000 description 1
- 108010009906 Angiopoietins Proteins 0.000 description 1
- 102400000068 Angiostatin Human genes 0.000 description 1
- 108010079709 Angiostatins Proteins 0.000 description 1
- 102000053640 Argininosuccinate synthases Human genes 0.000 description 1
- 108700024106 Argininosuccinate synthases Proteins 0.000 description 1
- 102000014461 Ataxins Human genes 0.000 description 1
- 108010078286 Ataxins Proteins 0.000 description 1
- 208000023275 Autoimmune disease Diseases 0.000 description 1
- 108010008014 B-Cell Maturation Antigen Proteins 0.000 description 1
- 102000006942 B-Cell Maturation Antigen Human genes 0.000 description 1
- 102100038080 B-cell receptor CD22 Human genes 0.000 description 1
- 102100024222 B-lymphocyte antigen CD19 Human genes 0.000 description 1
- 102100022005 B-lymphocyte antigen CD20 Human genes 0.000 description 1
- 108010074708 B7-H1 Antigen Proteins 0.000 description 1
- 102000006734 Beta-Globulins Human genes 0.000 description 1
- 108010087504 Beta-Globulins Proteins 0.000 description 1
- 101710124976 Beta-hexosaminidase A Proteins 0.000 description 1
- 108010039209 Blood Coagulation Factors Proteins 0.000 description 1
- 102000015081 Blood Coagulation Factors Human genes 0.000 description 1
- 102000007350 Bone Morphogenetic Proteins Human genes 0.000 description 1
- 108010007726 Bone Morphogenetic Proteins Proteins 0.000 description 1
- 108090000715 Brain-derived neurotrophic factor Proteins 0.000 description 1
- 102000004219 Brain-derived neurotrophic factor Human genes 0.000 description 1
- 102100035875 C-C chemokine receptor type 5 Human genes 0.000 description 1
- 101710149870 C-C chemokine receptor type 5 Proteins 0.000 description 1
- 102000014817 CACNA1A Human genes 0.000 description 1
- 102100024217 CAMPATH-1 antigen Human genes 0.000 description 1
- 102100031168 CCN family member 2 Human genes 0.000 description 1
- 102000017420 CD3 protein, epsilon/gamma/delta subunit Human genes 0.000 description 1
- 108050005493 CD3 protein, epsilon/gamma/delta subunit Proteins 0.000 description 1
- 102000049320 CD36 Human genes 0.000 description 1
- 108010045374 CD36 Antigens Proteins 0.000 description 1
- 101150013553 CD40 gene Proteins 0.000 description 1
- 108010065524 CD52 Antigen Proteins 0.000 description 1
- 102100025221 CD70 antigen Human genes 0.000 description 1
- 101100123850 Caenorhabditis elegans her-1 gene Proteins 0.000 description 1
- 108090000312 Calcium Channels Proteins 0.000 description 1
- 102000003922 Calcium Channels Human genes 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- 108010078791 Carrier Proteins Proteins 0.000 description 1
- 102000053642 Catalytic RNA Human genes 0.000 description 1
- 108090000994 Catalytic RNA Proteins 0.000 description 1
- 206010008025 Cerebellar ataxia Diseases 0.000 description 1
- 108010019670 Chimeric Antigen Receptors Proteins 0.000 description 1
- 102000011022 Chorionic Gonadotropin Human genes 0.000 description 1
- 108010062540 Chorionic Gonadotropin Proteins 0.000 description 1
- 108010005939 Ciliary Neurotrophic Factor Proteins 0.000 description 1
- 102100031614 Ciliary neurotrophic factor Human genes 0.000 description 1
- 102100022641 Coagulation factor IX Human genes 0.000 description 1
- 102100023804 Coagulation factor VII Human genes 0.000 description 1
- 206010053567 Coagulopathies Diseases 0.000 description 1
- 108091026890 Coding region Proteins 0.000 description 1
- 108010039419 Connective Tissue Growth Factor Proteins 0.000 description 1
- 241000711573 Coronaviridae Species 0.000 description 1
- 241000709687 Coxsackievirus Species 0.000 description 1
- 102000005927 Cysteine Proteases Human genes 0.000 description 1
- 108010005843 Cysteine Proteases Proteins 0.000 description 1
- 201000003883 Cystic fibrosis Diseases 0.000 description 1
- 108020004414 DNA Proteins 0.000 description 1
- 201000004624 Dermatitis Diseases 0.000 description 1
- 208000014094 Dystonic disease Diseases 0.000 description 1
- 201000011001 Ebola Hemorrhagic Fever Diseases 0.000 description 1
- 241001115402 Ebolavirus Species 0.000 description 1
- 241000196324 Embryophyta Species 0.000 description 1
- 102000005593 Endopeptidases Human genes 0.000 description 1
- 108010059378 Endopeptidases Proteins 0.000 description 1
- 241000709661 Enterovirus Species 0.000 description 1
- 241000991587 Enterovirus C Species 0.000 description 1
- 108010055196 EphA2 Receptor Proteins 0.000 description 1
- 108010055334 EphB2 Receptor Proteins 0.000 description 1
- 102100030340 Ephrin type-A receptor 2 Human genes 0.000 description 1
- 102100031968 Ephrin type-B receptor 2 Human genes 0.000 description 1
- 102400001368 Epidermal growth factor Human genes 0.000 description 1
- 101800003838 Epidermal growth factor Proteins 0.000 description 1
- 102000003951 Erythropoietin Human genes 0.000 description 1
- 108090000394 Erythropoietin Proteins 0.000 description 1
- 108700039887 Essential Genes Proteins 0.000 description 1
- 108010076282 Factor IX Proteins 0.000 description 1
- 108010023321 Factor VII Proteins 0.000 description 1
- 108010054218 Factor VIII Proteins 0.000 description 1
- 102000001690 Factor VIII Human genes 0.000 description 1
- 108010054265 Factor VIIa Proteins 0.000 description 1
- 108010014173 Factor X Proteins 0.000 description 1
- 108010071289 Factor XIII Proteins 0.000 description 1
- 102000003971 Fibroblast Growth Factor 1 Human genes 0.000 description 1
- 108090000386 Fibroblast Growth Factor 1 Proteins 0.000 description 1
- 102000003974 Fibroblast growth factor 2 Human genes 0.000 description 1
- 108090000379 Fibroblast growth factor 2 Proteins 0.000 description 1
- 102100035139 Folate receptor alpha Human genes 0.000 description 1
- 102000012673 Follicle Stimulating Hormone Human genes 0.000 description 1
- 108010079345 Follicle Stimulating Hormone Proteins 0.000 description 1
- 208000001914 Fragile X syndrome Diseases 0.000 description 1
- 102000003869 Frataxin Human genes 0.000 description 1
- 108090000217 Frataxin Proteins 0.000 description 1
- 102000034615 Glial cell line-derived neurotrophic factor Human genes 0.000 description 1
- 108091010837 Glial cell line-derived neurotrophic factor Proteins 0.000 description 1
- 101710088083 Glomulin Proteins 0.000 description 1
- 102400000321 Glucagon Human genes 0.000 description 1
- 108060003199 Glucagon Proteins 0.000 description 1
- 108010086800 Glucose-6-Phosphatase Proteins 0.000 description 1
- 102000003638 Glucose-6-Phosphatase Human genes 0.000 description 1
- 102100041003 Glutamate carboxypeptidase 2 Human genes 0.000 description 1
- 102100032530 Glypican-3 Human genes 0.000 description 1
- 102000004269 Granulocyte Colony-Stimulating Factor Human genes 0.000 description 1
- 108010017080 Granulocyte Colony-Stimulating Factor Proteins 0.000 description 1
- 108010051696 Growth Hormone Proteins 0.000 description 1
- 239000000095 Growth Hormone-Releasing Hormone Substances 0.000 description 1
- NYHBQMYGNKIUIF-UUOKFMHZSA-N Guanosine Chemical compound C1=NC=2C(=O)NC(N)=NC=2N1[C@@H]1O[C@H](CO)[C@@H](O)[C@H]1O NYHBQMYGNKIUIF-UUOKFMHZSA-N 0.000 description 1
- 108010078321 Guanylate Cyclase Proteins 0.000 description 1
- 102000014469 Guanylate cyclase Human genes 0.000 description 1
- 208000031886 HIV Infections Diseases 0.000 description 1
- 208000037357 HIV infectious disease Diseases 0.000 description 1
- 102100030595 HLA class II histocompatibility antigen gamma chain Human genes 0.000 description 1
- 208000005176 Hepatitis C Diseases 0.000 description 1
- 108090000100 Hepatocyte Growth Factor Proteins 0.000 description 1
- 102000003745 Hepatocyte Growth Factor Human genes 0.000 description 1
- 208000009889 Herpes Simplex Diseases 0.000 description 1
- 102000002268 Hexosaminidases Human genes 0.000 description 1
- 108010000540 Hexosaminidases Proteins 0.000 description 1
- 102100026122 High affinity immunoglobulin gamma Fc receptor I Human genes 0.000 description 1
- 101000833172 Homo sapiens AF4/FMR2 family member 2 Proteins 0.000 description 1
- 101000873082 Homo sapiens Ataxin-1 Proteins 0.000 description 1
- 101000895114 Homo sapiens Ataxin-2 Proteins 0.000 description 1
- 101000884305 Homo sapiens B-cell receptor CD22 Proteins 0.000 description 1
- 101000980825 Homo sapiens B-lymphocyte antigen CD19 Proteins 0.000 description 1
- 101000897405 Homo sapiens B-lymphocyte antigen CD20 Proteins 0.000 description 1
- 101000934356 Homo sapiens CD70 antigen Proteins 0.000 description 1
- 101001023230 Homo sapiens Folate receptor alpha Proteins 0.000 description 1
- 101000892862 Homo sapiens Glutamate carboxypeptidase 2 Proteins 0.000 description 1
- 101001014668 Homo sapiens Glypican-3 Proteins 0.000 description 1
- 101001082627 Homo sapiens HLA class II histocompatibility antigen gamma chain Proteins 0.000 description 1
- 101000913074 Homo sapiens High affinity immunoglobulin gamma Fc receptor I Proteins 0.000 description 1
- 101001057504 Homo sapiens Interferon-stimulated gene 20 kDa protein Proteins 0.000 description 1
- 101001055144 Homo sapiens Interleukin-2 receptor subunit alpha Proteins 0.000 description 1
- 101000917826 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor II-a Proteins 0.000 description 1
- 101000917824 Homo sapiens Low affinity immunoglobulin gamma Fc region receptor II-b Proteins 0.000 description 1
- 101000961414 Homo sapiens Membrane cofactor protein Proteins 0.000 description 1
- 101000623901 Homo sapiens Mucin-16 Proteins 0.000 description 1
- 101000934338 Homo sapiens Myeloid cell surface antigen CD33 Proteins 0.000 description 1
- 101000581981 Homo sapiens Neural cell adhesion molecule 1 Proteins 0.000 description 1
- 101000934346 Homo sapiens T-cell surface antigen CD2 Proteins 0.000 description 1
- 101000834948 Homo sapiens Tomoregulin-2 Proteins 0.000 description 1
- 101000662686 Homo sapiens Torsin-1A Proteins 0.000 description 1
- 101000801433 Homo sapiens Trophoblast glycoprotein Proteins 0.000 description 1
- 101000851376 Homo sapiens Tumor necrosis factor receptor superfamily member 8 Proteins 0.000 description 1
- 101000935117 Homo sapiens Voltage-dependent P/Q-type calcium channel subunit alpha-1A Proteins 0.000 description 1
- 101000760175 Homo sapiens Zinc finger protein 35 Proteins 0.000 description 1
- 108010091358 Hypoxanthine Phosphoribosyltransferase Proteins 0.000 description 1
- 102100029098 Hypoxanthine-guanine phosphoribosyltransferase Human genes 0.000 description 1
- 206010062016 Immunosuppression Diseases 0.000 description 1
- 241000712431 Influenza A virus Species 0.000 description 1
- 102100026878 Interleukin-2 receptor subunit alpha Human genes 0.000 description 1
- 108010063738 Interleukins Proteins 0.000 description 1
- 102000015696 Interleukins Human genes 0.000 description 1
- 208000027747 Kennedy disease Diseases 0.000 description 1
- 108010001831 LDL receptors Proteins 0.000 description 1
- 108090001090 Lectins Proteins 0.000 description 1
- 102000004856 Lectins Human genes 0.000 description 1
- 102000004058 Leukemia inhibitory factor Human genes 0.000 description 1
- 108090000581 Leukemia inhibitory factor Proteins 0.000 description 1
- 108010013563 Lipoprotein Lipase Proteins 0.000 description 1
- 102100022119 Lipoprotein lipase Human genes 0.000 description 1
- 102100029204 Low affinity immunoglobulin gamma Fc region receptor II-a Human genes 0.000 description 1
- 102100024640 Low-density lipoprotein receptor Human genes 0.000 description 1
- 102000009151 Luteinizing Hormone Human genes 0.000 description 1
- 108010073521 Luteinizing Hormone Proteins 0.000 description 1
- 208000015439 Lysosomal storage disease Diseases 0.000 description 1
- 102000043129 MHC class I family Human genes 0.000 description 1
- 108091054437 MHC class I family Proteins 0.000 description 1
- 102100024295 Maltase-glucoamylase Human genes 0.000 description 1
- 241000712079 Measles morbillivirus Species 0.000 description 1
- 102100039373 Membrane cofactor protein Human genes 0.000 description 1
- 208000036626 Mental retardation Diseases 0.000 description 1
- 108090000015 Mesothelin Proteins 0.000 description 1
- 102000003735 Mesothelin Human genes 0.000 description 1
- 102000014962 Monocyte Chemoattractant Proteins Human genes 0.000 description 1
- 108010064136 Monocyte Chemoattractant Proteins Proteins 0.000 description 1
- 102100023123 Mucin-16 Human genes 0.000 description 1
- 101100335081 Mus musculus Flt3 gene Proteins 0.000 description 1
- 101100369076 Mus musculus Tdgf1 gene Proteins 0.000 description 1
- 102100025243 Myeloid cell surface antigen CD33 Human genes 0.000 description 1
- 206010068871 Myotonic dystrophy Diseases 0.000 description 1
- 206010028885 Necrotising fasciitis Diseases 0.000 description 1
- 102000015336 Nerve Growth Factor Human genes 0.000 description 1
- 102000007072 Nerve Growth Factors Human genes 0.000 description 1
- 108010074223 Netrin-1 Proteins 0.000 description 1
- 102000009065 Netrin-1 Human genes 0.000 description 1
- 102100027347 Neural cell adhesion molecule 1 Human genes 0.000 description 1
- 206010029350 Neurotoxicity Diseases 0.000 description 1
- 102100029268 Neurotrophin-3 Human genes 0.000 description 1
- 108091034117 Oligonucleotide Proteins 0.000 description 1
- 102000007981 Ornithine carbamoyltransferase Human genes 0.000 description 1
- 101710198224 Ornithine carbamoyltransferase, mitochondrial Proteins 0.000 description 1
- 101150094724 PCSK9 gene Proteins 0.000 description 1
- 102000003982 Parathyroid hormone Human genes 0.000 description 1
- 108090000445 Parathyroid hormone Proteins 0.000 description 1
- 208000018737 Parkinson disease Diseases 0.000 description 1
- 235000019483 Peanut oil Nutrition 0.000 description 1
- 102000035195 Peptidases Human genes 0.000 description 1
- 108091005804 Peptidases Proteins 0.000 description 1
- 102000007079 Peptide Fragments Human genes 0.000 description 1
- 108010033276 Peptide Fragments Proteins 0.000 description 1
- 201000007100 Pharyngitis Diseases 0.000 description 1
- 108010038512 Platelet-Derived Growth Factor Proteins 0.000 description 1
- 102000010780 Platelet-Derived Growth Factor Human genes 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 229920001213 Polysorbate 20 Polymers 0.000 description 1
- 102100022019 Pregnancy-specific beta-1-glycoprotein 2 Human genes 0.000 description 1
- 102100024216 Programmed cell death 1 ligand 1 Human genes 0.000 description 1
- 102100023832 Prolyl endopeptidase FAP Human genes 0.000 description 1
- 239000004365 Protease Substances 0.000 description 1
- 101800004937 Protein C Proteins 0.000 description 1
- 241000125945 Protoparvovirus Species 0.000 description 1
- 108091030071 RNAI Proteins 0.000 description 1
- 241000725643 Respiratory syncytial virus Species 0.000 description 1
- 206010061603 Respiratory syncytial virus infection Diseases 0.000 description 1
- 208000007014 Retinitis pigmentosa Diseases 0.000 description 1
- 102100031176 Retinoid isomerohydrolase Human genes 0.000 description 1
- 108090000820 Rhodopsin Proteins 0.000 description 1
- 108091028664 Ribonucleotide Proteins 0.000 description 1
- 102220492384 Ribulose-phosphate 3-epimerase_T24S_mutation Human genes 0.000 description 1
- 102100036546 Salivary acidic proline-rich phosphoprotein 1/2 Human genes 0.000 description 1
- 101800001700 Saposin-D Proteins 0.000 description 1
- 241000710961 Semliki Forest virus Species 0.000 description 1
- 241000837158 Senecavirus A Species 0.000 description 1
- 206010040047 Sepsis Diseases 0.000 description 1
- 206010040070 Septic Shock Diseases 0.000 description 1
- 102100029014 Serine/threonine-protein phosphatase 2A 55 kDa regulatory subunit B beta isoform Human genes 0.000 description 1
- 101710109874 Serine/threonine-protein phosphatase 2A 55 kDa regulatory subunit B beta isoform Proteins 0.000 description 1
- 108091027967 Small hairpin RNA Proteins 0.000 description 1
- 108020004459 Small interfering RNA Proteins 0.000 description 1
- 102100022831 Somatoliberin Human genes 0.000 description 1
- 101710142969 Somatoliberin Proteins 0.000 description 1
- 102100038803 Somatotropin Human genes 0.000 description 1
- 108010019965 Spectrin Proteins 0.000 description 1
- 102000005890 Spectrin Human genes 0.000 description 1
- 108010061312 Sphingomyelin Phosphodiesterase Proteins 0.000 description 1
- 102000011971 Sphingomyelin Phosphodiesterase Human genes 0.000 description 1
- 208000009415 Spinocerebellar Ataxias Diseases 0.000 description 1
- 206010061372 Streptococcal infection Diseases 0.000 description 1
- 241000194048 Streptococcus equi Species 0.000 description 1
- 241001505901 Streptococcus sp. 'group A' Species 0.000 description 1
- QAOWNCQODCNURD-UHFFFAOYSA-N Sulfuric acid Chemical compound OS(O)(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-N 0.000 description 1
- 102100025237 T-cell surface antigen CD2 Human genes 0.000 description 1
- 102000006467 TATA-Box Binding Protein Human genes 0.000 description 1
- 108010044281 TATA-Box Binding Protein Proteins 0.000 description 1
- 108010041111 Thrombopoietin Proteins 0.000 description 1
- 102000036693 Thrombopoietin Human genes 0.000 description 1
- 102100026160 Tomoregulin-2 Human genes 0.000 description 1
- 102100037454 Torsin-1A Human genes 0.000 description 1
- 206010044221 Toxic encephalopathy Diseases 0.000 description 1
- 206010044248 Toxic shock syndrome Diseases 0.000 description 1
- 231100000650 Toxic shock syndrome Toxicity 0.000 description 1
- 102000004887 Transforming Growth Factor beta Human genes 0.000 description 1
- 108090001012 Transforming Growth Factor beta Proteins 0.000 description 1
- 102400001320 Transforming growth factor alpha Human genes 0.000 description 1
- 101800004564 Transforming growth factor alpha Proteins 0.000 description 1
- 108700019146 Transgenes Proteins 0.000 description 1
- 206010052779 Transplant rejections Diseases 0.000 description 1
- 102100031988 Tumor necrosis factor ligand superfamily member 6 Human genes 0.000 description 1
- 108050002568 Tumor necrosis factor ligand superfamily member 6 Proteins 0.000 description 1
- 102100040245 Tumor necrosis factor receptor superfamily member 5 Human genes 0.000 description 1
- 102100036857 Tumor necrosis factor receptor superfamily member 8 Human genes 0.000 description 1
- 102000014384 Type C Phospholipases Human genes 0.000 description 1
- 108010079194 Type C Phospholipases Proteins 0.000 description 1
- 108091000117 Tyrosine 3-Monooxygenase Proteins 0.000 description 1
- 102000048218 Tyrosine 3-monooxygenases Human genes 0.000 description 1
- 108010073929 Vascular Endothelial Growth Factor A Proteins 0.000 description 1
- 241000711975 Vesicular stomatitis virus Species 0.000 description 1
- 102100025330 Voltage-dependent P/Q-type calcium channel subunit alpha-1A Human genes 0.000 description 1
- 241000710886 West Nile virus Species 0.000 description 1
- 210000001766 X chromosome Anatomy 0.000 description 1
- 208000006269 X-Linked Bulbo-Spinal Atrophy Diseases 0.000 description 1
- 102100024672 Zinc finger protein 35 Human genes 0.000 description 1
- 238000002835 absorbance Methods 0.000 description 1
- 239000002253 acid Substances 0.000 description 1
- 230000002378 acidificating effect Effects 0.000 description 1
- 230000001154 acute effect Effects 0.000 description 1
- 108060000200 adenylate cyclase Proteins 0.000 description 1
- 102000030621 adenylate cyclase Human genes 0.000 description 1
- 102000005840 alpha-Galactosidase Human genes 0.000 description 1
- 108010030291 alpha-Galactosidase Proteins 0.000 description 1
- 108010028144 alpha-Glucosidases Proteins 0.000 description 1
- 108010080146 androgen receptors Proteins 0.000 description 1
- 125000000613 asparagine group Chemical group N[C@@H](CC(N)=O)C(=O)* 0.000 description 1
- FZCSTZYAHCUGEM-UHFFFAOYSA-N aspergillomarasmine B Natural products OC(=O)CNC(C(O)=O)CNC(C(O)=O)CC(O)=O FZCSTZYAHCUGEM-UHFFFAOYSA-N 0.000 description 1
- 201000004562 autosomal dominant cerebellar ataxia Diseases 0.000 description 1
- 230000001580 bacterial effect Effects 0.000 description 1
- 239000011324 bead Substances 0.000 description 1
- 230000008033 biological extinction Effects 0.000 description 1
- 229960002685 biotin Drugs 0.000 description 1
- 235000020958 biotin Nutrition 0.000 description 1
- 239000011616 biotin Substances 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- 229940112869 bone morphogenetic protein Drugs 0.000 description 1
- 229940077737 brain-derived neurotrophic factor Drugs 0.000 description 1
- 239000000872 buffer Substances 0.000 description 1
- 239000007853 buffer solution Substances 0.000 description 1
- 102220352428 c.92G>A Human genes 0.000 description 1
- 239000003593 chromogenic compound Substances 0.000 description 1
- 230000035602 clotting Effects 0.000 description 1
- 230000000295 complement effect Effects 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 230000003247 decreasing effect Effects 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 239000005549 deoxyribonucleoside Substances 0.000 description 1
- 239000005547 deoxyribonucleotide Substances 0.000 description 1
- 125000002637 deoxyribonucleotide group Chemical group 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 238000010790 dilution Methods 0.000 description 1
- 239000012895 dilution Substances 0.000 description 1
- 150000002009 diols Chemical class 0.000 description 1
- 229940042399 direct acting antivirals protease inhibitors Drugs 0.000 description 1
- 238000004090 dissolution Methods 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 229950002830 enadenotucirev Drugs 0.000 description 1
- 108060002566 ephrin Proteins 0.000 description 1
- 102000012803 ephrin Human genes 0.000 description 1
- 229940116977 epidermal growth factor Drugs 0.000 description 1
- 102000052116 epidermal growth factor receptor activity proteins Human genes 0.000 description 1
- 108700015053 epidermal growth factor receptor activity proteins Proteins 0.000 description 1
- 229940105423 erythropoietin Drugs 0.000 description 1
- 239000002329 esterase inhibitor Substances 0.000 description 1
- 210000003527 eukaryotic cell Anatomy 0.000 description 1
- 239000013613 expression plasmid Substances 0.000 description 1
- 229960004222 factor ix Drugs 0.000 description 1
- 229940012413 factor vii Drugs 0.000 description 1
- 229940012414 factor viia Drugs 0.000 description 1
- 229960000301 factor viii Drugs 0.000 description 1
- 229940012426 factor x Drugs 0.000 description 1
- 229940012444 factor xiii Drugs 0.000 description 1
- 108700014844 flt3 ligand Proteins 0.000 description 1
- 229940028334 follicle stimulating hormone Drugs 0.000 description 1
- 201000003415 fragile X-associated tremor/ataxia syndrome Diseases 0.000 description 1
- 210000001035 gastrointestinal tract Anatomy 0.000 description 1
- 230000009368 gene silencing by RNA Effects 0.000 description 1
- 238000010353 genetic engineering Methods 0.000 description 1
- MASNOZXLGMXCHN-ZLPAWPGGSA-N glucagon Chemical compound C([C@@H](C(=O)N[C@H](C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCSC)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H]([C@@H](C)O)C(O)=O)C(C)C)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](C)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CO)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CCCCN)NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@H](CC=1C=CC=CC=1)NC(=O)[C@@H](NC(=O)CNC(=O)[C@H](CCC(N)=O)NC(=O)[C@H](CO)NC(=O)[C@@H](N)CC=1NC=NC=1)[C@@H](C)O)[C@@H](C)O)C1=CC=CC=C1 MASNOZXLGMXCHN-ZLPAWPGGSA-N 0.000 description 1
- 229960004666 glucagon Drugs 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- 239000000122 growth hormone Substances 0.000 description 1
- RQFCJASXJCIDSX-UUOKFMHZSA-N guanosine 5'-monophosphate Chemical compound C1=2NC(N)=NC(=O)C=2N=CN1[C@@H]1O[C@H](COP(O)(O)=O)[C@@H](O)[C@H]1O RQFCJASXJCIDSX-UUOKFMHZSA-N 0.000 description 1
- 208000010710 hepatitis C virus infection Diseases 0.000 description 1
- 125000000487 histidyl group Chemical group [H]N([H])C(C(=O)O*)C([H])([H])C1=C([H])N([H])C([H])=N1 0.000 description 1
- 102000056417 human ATXN1 Human genes 0.000 description 1
- 102000056418 human ATXN2 Human genes 0.000 description 1
- 229940084986 human chorionic gonadotropin Drugs 0.000 description 1
- 208000033519 human immunodeficiency virus infectious disease Diseases 0.000 description 1
- 244000052637 human pathogen Species 0.000 description 1
- 230000005847 immunogenicity Effects 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 238000003780 insertion Methods 0.000 description 1
- 230000037431 insertion Effects 0.000 description 1
- 229940047124 interferons Drugs 0.000 description 1
- 229940047122 interleukins Drugs 0.000 description 1
- 238000001990 intravenous administration Methods 0.000 description 1
- 125000000741 isoleucyl group Chemical group [H]N([H])C(C(C([H])([H])[H])C([H])([H])C([H])([H])[H])C(=O)O* 0.000 description 1
- 101150044508 key gene Proteins 0.000 description 1
- 239000002523 lectin Substances 0.000 description 1
- 239000006193 liquid solution Substances 0.000 description 1
- 239000006194 liquid suspension Substances 0.000 description 1
- 229940040129 luteinizing hormone Drugs 0.000 description 1
- 125000003588 lysine group Chemical group [H]N([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])(N([H])[H])C(*)=O 0.000 description 1
- 230000002132 lysosomal effect Effects 0.000 description 1
- 210000004962 mammalian cell Anatomy 0.000 description 1
- 230000001404 mediated effect Effects 0.000 description 1
- 229910052751 metal Inorganic materials 0.000 description 1
- 239000002184 metal Substances 0.000 description 1
- 108091070501 miRNA Proteins 0.000 description 1
- 239000002679 microRNA Substances 0.000 description 1
- 239000002480 mineral oil Substances 0.000 description 1
- 235000010446 mineral oil Nutrition 0.000 description 1
- 208000009091 myxoma Diseases 0.000 description 1
- YOHYSYJDKVYCJI-UHFFFAOYSA-N n-[3-[[6-[3-(trifluoromethyl)anilino]pyrimidin-4-yl]amino]phenyl]cyclopropanecarboxamide Chemical compound FC(F)(F)C1=CC=CC(NC=2N=CN=C(NC=3C=C(NC(=O)C4CC4)C=CC=3)C=2)=C1 YOHYSYJDKVYCJI-UHFFFAOYSA-N 0.000 description 1
- 201000007970 necrotizing fasciitis Diseases 0.000 description 1
- 229940053128 nerve growth factor Drugs 0.000 description 1
- 108010081726 netrin-2 Proteins 0.000 description 1
- 230000007135 neurotoxicity Effects 0.000 description 1
- 231100000228 neurotoxicity Toxicity 0.000 description 1
- 229940056106 nipocalimab Drugs 0.000 description 1
- 229940121468 nirsevimab Drugs 0.000 description 1
- 238000005457 optimization Methods 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 239000003960 organic solvent Substances 0.000 description 1
- 239000006179 pH buffering agent Substances 0.000 description 1
- 229960001319 parathyroid hormone Drugs 0.000 description 1
- 239000000199 parathyroid hormone Substances 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 230000008506 pathogenesis Effects 0.000 description 1
- 239000000312 peanut oil Substances 0.000 description 1
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 1
- 239000003208 petroleum Substances 0.000 description 1
- PTMHPRAIXMAOOB-UHFFFAOYSA-L phosphoramidate Chemical compound NP([O-])([O-])=O PTMHPRAIXMAOOB-UHFFFAOYSA-L 0.000 description 1
- 230000004962 physiological condition Effects 0.000 description 1
- 229920001481 poly(stearyl methacrylate) Polymers 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 1
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 1
- 230000008092 positive effect Effects 0.000 description 1
- OXCMYAYHXIHQOA-UHFFFAOYSA-N potassium;[2-butyl-5-chloro-3-[[4-[2-(1,2,4-triaza-3-azanidacyclopenta-1,4-dien-5-yl)phenyl]phenyl]methyl]imidazol-4-yl]methanol Chemical compound [K+].CCCCC1=NC(Cl)=C(CO)N1CC1=CC=C(C=2C(=CC=CC=2)C2=N[N-]N=N2)C=C1 OXCMYAYHXIHQOA-UHFFFAOYSA-N 0.000 description 1
- 238000011533 pre-incubation Methods 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- 230000000750 progressive effect Effects 0.000 description 1
- 210000001236 prokaryotic cell Anatomy 0.000 description 1
- 230000001681 protective effect Effects 0.000 description 1
- 229960000856 protein c Drugs 0.000 description 1
- 239000002994 raw material Substances 0.000 description 1
- 230000006798 recombination Effects 0.000 description 1
- 238000005215 recombination Methods 0.000 description 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 230000010076 replication Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 208000030925 respiratory syncytial virus infectious disease Diseases 0.000 description 1
- 210000002345 respiratory system Anatomy 0.000 description 1
- 108010054126 retinoid isomerohydrolase Proteins 0.000 description 1
- 239000002342 ribonucleoside Substances 0.000 description 1
- 239000002336 ribonucleotide Substances 0.000 description 1
- 125000002652 ribonucleotide group Chemical group 0.000 description 1
- 108091092562 ribozyme Proteins 0.000 description 1
- 229950005039 rozanolixizumab Drugs 0.000 description 1
- 102220221396 rs1060500635 Human genes 0.000 description 1
- 239000000523 sample Substances 0.000 description 1
- 238000012163 sequencing technique Methods 0.000 description 1
- 239000004055 small Interfering RNA Substances 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 239000003549 soybean oil Substances 0.000 description 1
- 235000012424 soybean oil Nutrition 0.000 description 1
- 210000000130 stem cell Anatomy 0.000 description 1
- 210000002536 stromal cell Anatomy 0.000 description 1
- 239000013595 supernatant sample Substances 0.000 description 1
- 239000004094 surface-active agent Substances 0.000 description 1
- 238000013268 sustained release Methods 0.000 description 1
- 239000012730 sustained-release form Substances 0.000 description 1
- 239000006188 syrup Substances 0.000 description 1
- 235000020357 syrup Nutrition 0.000 description 1
- 101150047061 tag-72 gene Proteins 0.000 description 1
- ZRKFYGHZFMAOKI-QMGMOQQFSA-N tgfbeta Chemical compound C([C@H](NC(=O)[C@H](C(C)C)NC(=O)CNC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CC(C)C)NC(=O)CNC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](NC(=O)[C@H](C)NC(=O)[C@H](C)NC(=O)[C@@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](N)CCSC)C(C)C)[C@@H](C)CC)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](C)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](C)C(=O)N[C@@H](CC(C)C)C(=O)N1[C@@H](CCC1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(C)C)C(O)=O)C1=CC=C(O)C=C1 ZRKFYGHZFMAOKI-QMGMOQQFSA-N 0.000 description 1
- RYYWUUFWQRZTIU-UHFFFAOYSA-K thiophosphate Chemical compound [O-]P([O-])([O-])=S RYYWUUFWQRZTIU-UHFFFAOYSA-K 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 238000010361 transduction Methods 0.000 description 1
- 230000026683 transduction Effects 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 238000013519 translation Methods 0.000 description 1
- 238000002054 transplantation Methods 0.000 description 1
- 239000002753 trypsin inhibitor Substances 0.000 description 1
- 210000004881 tumor cell Anatomy 0.000 description 1
- 238000000108 ultra-filtration Methods 0.000 description 1
- 238000011144 upstream manufacturing Methods 0.000 description 1
- VBEQCZHXXJYVRD-GACYYNSASA-N uroanthelone Chemical compound C([C@@H](C(=O)N[C@H](C(=O)N[C@@H](CS)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CS)C(=O)N[C@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)NCC(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H](CO)C(=O)NCC(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CS)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(O)=O)C(C)C)[C@@H](C)O)NC(=O)[C@H](CO)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@@H](NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H](CCSC)NC(=O)[C@H](CS)NC(=O)[C@@H](NC(=O)CNC(=O)CNC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CS)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)CNC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@H](CO)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CS)NC(=O)CNC(=O)[C@H]1N(CCC1)C(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@@H](N)CC(N)=O)C(C)C)[C@@H](C)CC)C1=CC=C(O)C=C1 VBEQCZHXXJYVRD-GACYYNSASA-N 0.000 description 1
- 108700026220 vif Genes Proteins 0.000 description 1
- 230000009385 viral infection Effects 0.000 description 1
- 230000003612 virological effect Effects 0.000 description 1
- 230000007923 virulence factor Effects 0.000 description 1
- 239000000304 virulence factor Substances 0.000 description 1
- 108010025625 vocimagene amiretrorepvec Proteins 0.000 description 1
- 108010078375 voltage-dependent calcium channel (P-Q type) Proteins 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
Images
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N9/00—Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
- C12N9/14—Hydrolases (3)
- C12N9/48—Hydrolases (3) acting on peptide bonds (3.4)
- C12N9/50—Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
- C12N9/52—Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from bacteria or Archaea
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/43—Enzymes; Proenzymes; Derivatives thereof
- A61K38/46—Hydrolases (3)
- A61K38/48—Hydrolases (3) acting on peptide bonds (3.4)
- A61K38/4873—Cysteine endopeptidases (3.4.22), e.g. stem bromelain, papain, ficin, cathepsin H
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/66—Microorganisms or materials therefrom
- A61K35/76—Viruses; Subviral particles; Bacteriophages
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
- A61K39/39533—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
- A61K39/3955—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N9/00—Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
- C12N9/14—Hydrolases (3)
- C12N9/48—Hydrolases (3) acting on peptide bonds (3.4)
- C12N9/50—Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
- C12N9/52—Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from bacteria or Archaea
- C12N9/54—Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from bacteria or Archaea bacteria being Bacillus
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12R—INDEXING SCHEME ASSOCIATED WITH SUBCLASSES C12C - C12Q, RELATING TO MICROORGANISMS
- C12R2001/00—Microorganisms ; Processes using microorganisms
- C12R2001/01—Bacteria or Actinomycetales ; using bacteria or Actinomycetales
- C12R2001/46—Streptococcus ; Enterococcus; Lactococcus
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Y—ENZYMES
- C12Y304/00—Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
- C12Y304/22—Cysteine endopeptidases (3.4.22)
- C12Y304/2201—Streptopain (3.4.22.10)
Definitions
- the present invention relates to the field of biotechnology, in particular to a mutant of an immunoglobulin-degrading enzyme.
- Streptococcus pyogenes is one of the common pathogens in humans and animals. It is widely found in nature and in the oropharyngeal cavity, respiratory tract and intestinal tract of humans or animals. Streptococcal infection may induce related diseases, from milder diseases such as pyogenic dermatitis and pharyngitis, to more serious diseases such as sepsis, necrotizing fasciitis and toxic shock syndrome.
- Immunoglobulin G-degrading enzyme of Streptococcus pyogenes (IdeS), a common Group A Streptococcus pyogenes (GAS) cysteine protease, has endopeptidase activity that hydrolyzes IgG (Agniswamy J, Lei B, Musser J M et al., J Biol Chem, 2004, 279: 52789-52796.
- Immunoglobulin G is the main antibody component in serum, and accounts for about 75% of serum immunoglobulins. It mainly plays a protective role in the body's immunity and can effectively prevent infectious diseases. In addition to its protective effect, IgG is also related to diseases. In some autoimmune diseases, anti-IgG antibodies react with the body's own molecules, and IgG may cause acute transplant rejection in organ transplantation. IdeS specifically degrades IgG, causing IgG to lose its inherent function, thus achieving immunosuppression.
- IdeS used in clinical practice has the problems of poor activity and high pre-existing antibodies in the human body.
- IdeS is a virulence factor of human pathogens.
- Clinical studies have found that anti-IdeS antibodies are detected nearly 100% in normal people under normal physiological conditions, which leads to low administration efficiency and safety concerns in the use of IdeS.
- Protease IdeE with a sequence homology of about 70% to IdeS is derived from a Streptococcus equi subsp. zooepidemicus—Streptococcus equi ssp. equi, an equine pathogen (Jonas Lannerg ⁇ rd, Bengt Guss. FEMS Microbiol Lett, 2006, 262: 230-235).
- IdeE is derived from an equine pathogen, it is speculated that its pre-existing antibodies in the human body may be much lower than IdeS, and it is more suitable for the development of immunosuppressants for treating and preventing diseases mediated by anti-IgG antibodies.
- wild-type IdeE like IdeS, also has the problem of low activity. Therefore, it is necessary to improve the activity of IdeE through molecular design and mutation screening, reducing doses used in clinical practice, thus reducing the risks posed by high doses of proteins of bacterial origin. This is exactly what the present invention aims to achieve.
- the first aspect of the present invention relates to a mutant of an immunoglobulin-degrading enzyme IdeE, wherein the immunoglobulin-degrading enzyme IdeE comprises or consists of an amino acid sequence as set forth in SEQ ID NO: 2 in the Sequence Listing; and the mutant comprises a mutation selected from the group consisting of:
- the second aspect of the present invention relates to a protein comprising the mutant of the present invention.
- the protein is linked to a secretory signal sequence and/or methionine at the N-terminus of the mutant; and/or the protein is linked to a histidine tag at the C-terminus of the mutant.
- the third to fifth aspects of the present invention relate to a nucleotide encoding the mutant or protein of the present invention, an expression vector comprising the nucleotide, and a host cell comprising the expression vector or expressing the mutant or protein of the present invention.
- the sixth aspect of the present invention relates to a composition or kit, comprising: the mutant or protein of the present invention; and optionally, a substance selected from the group consisting of: a pharmaceutically acceptable carrier or excipient, an antibody or an Fc-containing protein and a viral vector drug.
- FIG. 8 shows the SDS-PAGE gel electrophoresis pattern of cleavage products resulting from cleavage of human IgG1 by mutant E97D_del18 and IdeS at different concentrations.
- FIG. 9 shows the SDS-PAGE gel electrophoresis pattern of cleavage products resulting from cleavage of human IgG1 by mutant E97D_del18 and IdeZ at different concentrations.
- FIG. 10 shows the SDS-PAGE gel electrophoresis pattern of cleavage products resulting from cleavage of human IVIg by mutant E97D_del18 in the sera and plasmas of mice.
- FIG. 11 shows the SDS-PAGE gel electrophoresis pattern of cleavage products produced by mutant E97D_del18 in the sera of mice and humans.
- FIG. 12 A- 12 D show the SDS-PAGE gel electrophoresis patterns of cleavage products resulting from cleavage of IgG by E97D_del18 at different concentrations in the sera of beagles, rats, mice, rabbits, monkeys and pigs.
- FIG. 13 shows the SDS-PAGE gel electrophoresis pattern of cleavage products resulting from cleavage of human IVIg by E97D_del18 at different times in mice.
- a functional polypeptide with the activity of immunoglobulin-degrading enzyme comprising a mutant based on an amino acid sequence as set forth in SEQ ID NO: 2, wherein the mutant is selected from the group consisting of:
- the mutant of the present invention has the function of the immunoglobulin-degrading enzyme IdeE, and preferably further has improved IgG-cleaving activity and thermal stability.
- the term “having higher activity than that of the immunoglobulin-degrading enzyme IdeE” in the present invention refers to the ability of the mutant to degrade immunoglobulins over that of the wild-type immunoglobulin-degrading enzyme IdeE.
- the term “having higher thermal stability than that of IdeE” in the present invention refers to the ability of the mutant after being maintained at a certain temperature for a period of time to degrade immunoglobulins over that of the wild-type immunoglobulin-degrading enzyme IdeE under the same conditions.
- the mutant of the present invention is preferably produced by means of genetic engineering recombination.
- the mutant has at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity to the sequence as set forth in SEQ ID NO: 2.
- amino acid at position 8, 10, 24, 59, 97 or 280 is substituted, for example, the amino acid sequence of the resulting mutant is as set forth in any one of SEQ ID NOs: 3-17 and SEQ ID NO: 35;
- amino acid substitution is selected from the group consisting of:
- amino acid substitution is selected from the group consisting of:
- the combinatorial mutation is further performed based on the three mutants of SEQ ID NOs:14-16, and the amino acid sequence of the resulting mutant is as set forth in SEQ ID NO: 35 in the Sequence Listing.
- the amino acid sequence of the resulting mutant is as set forth in SEQ ID NO: 35 in the Sequence Listing.
- the first 18 amino acids at their N-termini are further deleted, and the amino acid sequences of the resulting mutants are as set forth in SEQ ID NOs: 25-29 in the Sequence Listing.
- the mutant of the present invention can also be further mutated, and the sequence of the variant obtained through further mutation has at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity to the sequence of SEQ ID NO: 2, and the variant also has the function of the immunoglobulin-degrading enzyme IdeE.
- IdeE immunoglobulin-degrading enzyme
- the complete sequence of the IdeE used in the present invention is publicly available under the GenBank accession number ABF57910.1, and its sequence is provided herein as SEQ ID NO: 1.
- the sequence comprises an N-terminal methionine, followed by a 33-amino acid secretory signal sequence, followed by an IdeE coding sequence.
- the N-terminal methionine and the signal sequence are usually removed to form a mature IdeE protein, the sequence of which is provided herein as SEQ ID NO: 2.
- all references to the numbers of the amino acid positions in the sequences of the immunoglobulin-degrading enzymes disclosed herein are based on the numbers of the corresponding positions in SEQ ID NO: 2 starting from the N-terminus.
- the present invention also provides a protein comprising the mutant as described above.
- the protein comprises a signal peptide at the N-terminus of the mutant described above; preferably, the protein is linked to a secretory signal sequence at the N-terminus of the mutant, and to methionine at the N-terminus of the secretory sequence, and/or is linked to a histidine tag at the C-terminus of the mutant; more preferably, the protein comprises or consists of the following from the N-terminus to the C-terminus: methionine, the secretory signal sequence and the mutant.
- nucleotide encoding the protein or mutant as described above.
- the present invention also provides an expression vector comprising the nucleotide.
- the present invention also provides a host cell comprising the expression vector as described above, or a host cell expressing the protein or mutant as described above.
- the host cell can be a conventional cell for expressing proteins or polypeptides in the art, and is selected from E. coli cells, yeast cells, etc.
- a composition comprising an immunoglobulin-degrading enzyme or a mutant thereof or a protein comprising an immunoglobulin-degrading enzyme or a mutant thereof; and optionally, a pharmaceutically acceptable carrier or excipient.
- the immunoglobulin-degrading enzyme is selected from IdeE, IdeS and IdeZ.
- the mutant of the immunoglobulin-degrading enzyme is a mutant as described above, and the protein is a protein comprising the mutant as described above.
- the composition of the present invention further comprises: an antibody or an Fc-containing protein.
- the antibody target is selected from the group consisting of: a cell surface protein, a cytokine, a hormone, an enzyme, an intracellular messenger, an intercellular messenger and an immune checkpoint.
- the composition of the present invention further comprises: a viral vector drug, preferably selected from the group consisting of: an oncolytic virus, a gene therapy virus and a viral vector vaccine.
- the composition of the present invention further comprises: an agent capable of reducing the IgG level in the blood, preferably selected from the group consisting of: an FcRn antibody and an Fc fragment variant with a high affinity to FcRn.
- the antibody target can be a cell surface protein, including but not limited: AFP, ⁇ v integrin, ⁇ 4 ⁇ 7 integrin, BCMA, CD2, CD3, CD19, CD20, CD22, CD25, CD30, CD32, CD33, CD36, CD40, CD46, CD52, CD56, CD64, CD70, CD74, CD79, CD80, CD86, CD105, CD121, CD123, CD133, CD138, CD174, CD205, CD227, CD326, CD340, CEA, c-Met, Cripto, CA1X, Claudin18.2, ED-B, EGFR, EpCAM, EphA2, EphB2, FAP, FOLR1, GD2, Globo H, GPC3, GPNMB, HER-1, HER-2, HER-3, MAGE-A3, mesothelin, MUC16, GPNMB, PSMA, TMEFF2, TAG-72, 5T4, ROR-1, Sca-1, Sca-1, Sca-1, Sc
- the antibody target can be a cytokine, including but not limited to: interleukins IL-1 to IL-13, tumor necrosis factors ⁇ and ⁇ , interferons ⁇ , ⁇ and ⁇ , tumor growth factor ⁇ (TGF- ⁇ ) colony stimulating factor (CSF) or granulocyte-monocyte colony stimulating factor (GM-CSF).
- TGF- ⁇ tumor growth factor ⁇
- CSF colony stimulating factor
- GM-CSF granulocyte-monocyte colony stimulating factor
- the antibody target can be a hormone, an enzyme, and an intracellular and intercellular messenger, such as adenylate cyclase, guanylate cyclase or phospholipase C.
- the antibody target can be an immune checkpoint, including: CTLA-4, PD-1, PD-L1, TIM-3, LAG3, Siglec15, 4-1BB, GITR, OX40, CD40L, CD28, TIGIT and VISTA.
- an immune checkpoint including: CTLA-4, PD-1, PD-L1, TIM-3, LAG3, Siglec15, 4-1BB, GITR, OX40, CD40L, CD28, TIGIT and VISTA.
- the combination further comprises a targeted drug or a chemotherapeutic drug or an immune checkpoint blocker
- the targeted drug is selected from an epigenetic drug, such as a histone deacetylase inhibitor, an inhibitor targeting the PI3K/Akt/mTOR signaling pathway, such as Tricibine, and a tyrosine kinase inhibitor, such as sunitinib
- the chemotherapeutic drug is selected from an immunosuppressant, such as cyclophosphamide, thalidomide and pomalidomide, a proteasome inhibitor, such as bortezomib, a cytotoxic drug, such as gemcitabine and temozolomide, and a cell cycle non-specific drug, such as mitoxantrone
- the immune checkpoint blocker is selected from an anti-CTLA-4 antibody, an anti-PD-1 antibody, an anti-TIM-3 antibody, an anti-LAG3 antibody, an anti-Siglec15 antibody, an epigenetic drug, such as a his
- the polypeptide drug capable of reducing the IgG level in the blood can block the binding of IgG in the blood to the FcRn protein.
- the affinity of the polypeptide to human FcRn protein is higher than that of IgG in the blood to human FcRn protein; and the IgG is selected from IgG1, IgG2, IgG3 and IgG4.
- the polypeptide comprises an antibody Fc fragment variant, which comprises a mutation capable of increasing the affinity of Fc to FcRn, preferably YTE, YTEKF, LS or NHS, wherein the antibody Fc fragment is, for example, Efgartigimod.
- the variant can be a monomer, a dimer or a multimer.
- the positions of the mutations of YTE, YTEKF, LS, NHS, etc., which can be used in the present invention, are as described by Dall'Acqua et al. (WF, D. A. et al. (2002). Journal of immunology (Baltimore, Md.: 1950) 169(9): 5171-5180.) and Lee et al. (Lee, C. H. et al. (2019). Nat Commun 10(1): 5031.), respectively.
- the mutation is performed on one selected from human IgG, which is selected from IgG1, IgG2, IgG3 and IgG4.
- Fc fragment variants which can be used in the present invention, comprise mutations including but not limited to those as described by Dall'Acqua et al. (WF, D. A. et al. (2002). Journal of immunology (Baltimore, Md.: 1950) 169(9): 5171-5180.), Shan et al. (Shan, L. et al. (2016). PLoS One 11(8): e0160345.), Lee et al. (Lee, C. H. et al. (2019). Nat Commun 10(1): 5031.), Mackness et al. (Mackness, B. C. et al. (2019). MAbs 11(7): 1276-1288.) and Christophe et al. (Dumet Christophe, Pottier Jaremy, Gouilleux-Gruart Valérie et al., MAbs, 2019, 11: 1341-1350.).
- the polypeptide comprises an antibody Fc fragment variant, which comprises a mutation capable of increasing the affinity of Fc to Fc ⁇ R, preferably S239D/I322E, S239D/I322E/A330L, K326W/E333S or R214K mutation, preferably afucosylation modification.
- the variant can be a monomer, a dimer or a multimer.
- Other Fc fragment variants, which can be used in the present invention comprise mutations including but not limited to those as described by Wang et al. (Wang Xinhua., Mathieu Mary., Brezski Randall J. (2016). Protein Cell, 9(1), 63-73. doi: 10.1007/s13238-017-0473-8).
- the variant comprising a mutation capable of increasing the affinity of Fc to FcRn also comprises a mutation capable of increasing the affinity of Fc to Fc ⁇ R.
- the variant can be a monomer, a dimer or a multimer.
- the polypeptide is selected from an anti-FcRn antibody, such as Nipocalimab, Rozanolixizumab, RVT-1401, HBM9161, ALXN1830, SYNT001 or Nirsevimab.
- an anti-FcRn antibody such as Nipocalimab, Rozanolixizumab, RVT-1401, HBM9161, ALXN1830, SYNT001 or Nirsevimab.
- the polypeptide is selected from a small peptide fragment capable of specifically binding to FcRn and with a length of 10-70 amino acids, such as ABY-039.
- the polypeptide is selected from an Fc multimer capable of specifically binding to FcRn, such as GL-2045, M230, PRIM, HexaGardTM, CSL777 or hexavalent molecules by UCB.
- FcRn Fc multimer capable of specifically binding to FcRn, such as GL-2045, M230, PRIM, HexaGardTM, CSL777 or hexavalent molecules by UCB.
- the polypeptide includes, but is not limited to, a polypeptide fragment as described by Sockolosky et al. (Sockolosky Jonathan T, Szoka Francis C. Adv. Drug Deliv. Rev., 2015, 91: 109-24).
- the virus used in the viral vector drug is selected from an ssDNA virus, a dsDNA virus, an ssRNA virus or a dsRNA virus; and/or the virus used in the viral vector drug is selected from a wild-type virus strain or naturally attenuated strain, a genetically engineered and selective attenuated strain, a gene-loaded virus strain and a gene transcription-targeting virus strain.
- the wild-type virus strain or naturally attenuated strain is selected from Newcastle disease virus, reovirus, mumps virus, West Nile virus, adenovirus, vaccinia virus, etc.
- the genetically engineered and selective attenuated strain enables the virus to selectively replicate in a tumor by manually deleting a key gene, such as thymidine kinase (TK)-knockout genetically modified human herpes simplex virus I (HSV-1), and the genetically engineered and selective attenuated strain is, for example, ONYX-015 or G207.
- ONYX-015 a segment of 827 bp in the E1b region is deleted, and a point mutation is made at the gene for protein E1B55K, so that its expression is terminated prematurely, and the E1B55K protein can not be expressed.
- G207 the ⁇ 34.5 gene is deleted, which is the determinant of HSV-1 neurotoxicity.
- the gene-loaded virus strain is loaded with an exogenous gene, such as one of granulocyte-macrophage colony stimulating factor (GM-CSF), and the gene-loaded virus strain is, for example, JX-594 or T-VEC.
- GM-CSF granulocyte-macrophage colony stimulating factor
- the gene transcription-targeting virus strain enables to control the replication of the oncolytic virus in a tumor cell by inserting a tissue- or tumor-specific promoter upstream of an essential gene of the virus, and the gene transcription-targeting virus strain is, for example, G92A.
- the ssDNA virus is selected from parvovirus, preferably H-1PV virus.
- the dsDNA virus is selected from herpes simplex virus, adenovirus and poxvirus; more preferably, the herpes simplex virus is preferably herpes simplex virus type I (HSV-1), such as R3616, T-VEC, HF10, G207, NV1020 and OrienX010; the poxvirus is selected from Pexa-Vec (a vaccinia virus), JX-594 (a vaccinia virus), GL-ONC1 and Myxoma; and the adenovirus is selected from Enadenotucirev, DNX-2401, C-REV, NG-348, ProsAtak, CG0070, ADV-TK, EDS01, KH901, H101, H103, VCN-01 and Telomelysin (OBP-301).
- HSV-1 herpes simplex virus type I
- the poxvirus is selected from Pexa-Vec (a vaccinia virus), JX-594 (a vaccinia
- the ssRNA virus is selected from picornavirus, alphavirus, retrovirus, paramyxovirus and rhabdovirus; preferably, the picornavirus is selected from CAVATAK, PVS-RIPO, CVA21 (an enterovirus) and RIGVIR, the alphavirus is selected from M1, Sindbis AR339 and Semliki Forest virus, the retrovirus is selected from Toca511, the paramyxovirus is selected from MV-NIS and PV701 (a Newcastle disease virus), and the rhabdovirus is selected from VSV-IFN ⁇ , MG1-MAGEA3 and VSV-GP.
- the picornavirus is selected from CAVATAK, PVS-RIPO, CVA21 (an enterovirus) and RIGVIR
- the alphavirus is selected from M1, Sindbis AR339 and Semliki Forest virus
- the retrovirus is selected from Toca511
- the paramyxovirus is selected from MV-NIS and PV701 (a Newcastle disease virus
- the dsRNA virus is selected from reovirus; preferably, the reovirus is selected from Pelareorep, Reolysin, vaccinia virus, mumps virus and human immunodeficiency virus (HIV).
- the RNA virus is selected from reovirus, coxsackievirus, poliovirus, porcine Seneca Valley virus, measles virus, Newcastle disease virus, vesicular stomatitis virus and influenza virus.
- the oncolytic virus expresses an exogenous gene, preferably those of a Bispecific T cell engager (BiTE), an scFv fragment, a cytokine and a chemokine.
- the BiTE can bind to a molecule that activates T cells such as CD 3, and can also bind to an antigen target on the surface of a cancer cell.
- the scFv targets an immune checkpoint, including CTLA-4, PD-1, TIM-3, LAG3, Siglec15, 4-1BB, GITR, OX40, CD40L, CD28, TIGIT and VISTA.
- the cytokine and chemokine are, for example, GM-CSF, interleukin-2 (IL-2), interleukin-12 (IL-12), an interferon (IFN), a tumor necrosis factor (TNF), soluble CD80 and CCL3.
- IL-2 interleukin-2
- IL-12 interleukin-12
- IFN interferon
- TNF tumor necrosis factor
- the gene therapy virus expresses an exogenous gene, which encodes a protein required for a gene-deficient disease selected from acidic ⁇ -glucosidase, copper-transporting ATPase 2, ⁇ -galactosidase, argininosuccinate synthase, ⁇ -glucocerebrosidase, ⁇ -hexosaminidase A, Cl protease inhibitors or Cl esterase inhibitors, glucose 6-phosphatase, insulin, glucagon, growth hormone, parathyroid hormone, growth hormone releasing factor, follicle stimulating hormone, luteinizing hormone, human chorionic gonadotropin, vascular endothelial growth factor, angiopoietin, angiostatin, granulocyte colony-stimulating factor, erythropoietin, connective tissue growth factor, basic fibroblast growth factor, acidic fibroblast growth factor, epidermal growth factor, transforming growth factor
- the gene therapy virus carries an exogenous gene, which encodes an inhibitory nucleic acid selected from siRNA, antisense molecules, miRNA, RNAi, ribozymes and shRNA.
- the inhibitory nucleic acid binds to a polynucleotide repeat disease-related gene, the transcript of which or the polynucleotide of the transcript of which is repeated.
- the disease-related gene encodes a related protein selected from huntington protein (HTT), androgen receptor on the X chromosome in spinobulbar muscular atrophy, human Ataxin-1/-2/-3/-7, Cav2.1 P/Q voltage-dependent calcium channel (CACNA1A), TATA-binding protein, Ataxin8 opposite strand (ATXN80S), serine/threonine protein phosphatase 2A 55 kDa regulatory subunit B beta isoform in spinocerebellar ataxia (types 1, 2, 3, 6, 7, 8, 12 and 17), FMR1 (Fragile X Mental Retardation 1) in fragile X syndrome, FMR1 (Fragile X Mental Retardation 1) in fragile X-associated tremor/ataxia syndrome, FMR1 (Fragile X Mental Retardation 2) or AF4/FMR2 family member 2 in fragile XE mental retardation, myotonin-protein kinase (MT-PK) in myotonic dystrophy and
- the disease-related gene is selected from a mutant of superoxide dismutase 1 (SOD1) gene, a gene related to pathogenesis of Parkinson's disease and/or Alzheimer's disease, apolipoprotein B (APOB) gene, PCSK9 gene, HIV infection-related genes (HIVTat, TAR, HIVTAR and CCR5), influenza A virus genome/gene sequences in influenza virus infection, severe acute respiratory syndrome (SARS) coronavirus genome/gene sequences in SARS infection, respiratory syncytial virus genome/gene sequences in respiratory syncytial virus infection, Ebola virus genome/gene sequences in Ebola virus infection, hepatitis B and C virus genome/gene sequences in hepatitis B and C virus infection, herpes simplex virus (HSV) genome/gene sequences in HSV infection, coxsackievirus B3 genome/gene sequences in coxsackievirus B3 infection, silencing of a pathogenic allele of a gene (allele-
- the present invention also provides an article of manufacture, comprising the mutant or protein as described above; and a therapeutic agent selected from a viral vector drug, an antibody, and a polypeptide drug capable of reducing the IgG level in the blood.
- the present invention also provides a kit or kit of parts, comprising: 1) a therapeutically effective amount of an agent, including the mutant as described above; and 2) a therapeutically effective amount of a therapeutic agent selected from a viral vector drug, an antibody, and a polypeptide drug capable of reducing the IgG level in the blood; preferably, the viral vector drug is an oncolytic virus or a gene therapy virus.
- the kit can further comprise 3) a targeted drug or a chemotherapeutic drug or an immune checkpoint blocker.
- the targeted drug is selected from an epigenetic drug, such as a histone deacetylase inhibitor, an inhibitor targeting the PI3K/Akt/mTOR signaling pathway, such as Tricibine, and a tyrosine kinase inhibitor, such as sunitinib;
- the chemotherapeutic drug is selected from an immunosuppressant, such as cyclophosphamide, thalidomide and pomalidomide, a proteasome inhibitor, such as bortezomib, a cytotoxic drug, such as gemcitabine and temozolomide, and a cell cycle non-specific drug, such as mitoxantrone;
- the immune checkpoint blocker is selected from an anti-CTLA-4 antibody, an anti-PD-1 antibody, an anti-TIM-3 antibody, an anti-LAG3 antibody, an anti-Siglec15 antibody, an anti-4-1BB antibody, an anti-GITR antibody, an anti-OX40 antibody, an anti-CD40L antibody, an anti-CD28
- the kit or kit of parts comprises a part A, comprising a therapeutically effective amount of the mutant or protein as described above; and a part B, comprising a therapeutically effective amount of a therapeutic agent selected from a viral vector drug, preferably an oncolytic virus or a gene therapy virus, an antibody, and a polypeptide drug capable of reducing the IgG level in the blood.
- the kit of parts can further comprise a part C.
- the part C comprises a targeted drug or a chemotherapeutic drug or an immune checkpoint blocker.
- the targeted drug is selected from an epigenetic drug, such as a histone deacetylase inhibitor, an inhibitor targeting the PI3K/Akt/mTOR signaling pathway, such as Tricibine, and a tyrosine kinase inhibitor, such as sunitinib;
- the chemotherapeutic drug is selected from an immunosuppressant, such as cyclophosphamide, thalidomide and pomalidomide, a proteasome inhibitor, such as bortezomib, a cytotoxic drug, such as gemcitabine and temozolomide, and a cell cycle non-specific drug, such as mitoxantrone;
- the immune checkpoint blocker is selected from an anti-CTLA-4 antibody, an anti-PD-1 antibody, an anti-TIM-3 antibody, an anti-LAG3 antibody, an anti-Siglec15 antibody, an anti-4-1BB antibody, an anti-GITR antibody, an anti-OX40 antibody, an anti-CD40L antibody, an anti-CD28
- the kit can comprise instructions on the administration of the therapeutically effective amount of the mutant or protein as described above and the therapeutically effective amount of the therapeutic agent (e.g., dose information and administration interval information).
- the therapeutic agent is selected from a viral vector drug, preferably an oncolytic virus or a gene therapy virus, an antibody, and a polypeptide drug capable of reducing the IgG level in the blood.
- Some examples of methods include the use of mammalian cell expression systems to produce viral particles, such as the use of HEK293 cells to produce adenovirus viral vector drugs (Freedman Joshua D, Duffy Margaret R, Lei-Rossmann Janet et al., An Oncolytic Virus Expressing a T-cell Engager Simultaneously Targets Cancer and Immunosuppressive Stromal Cells. [J]. Cancer Res., 2018, 78: 6852-6865).
- the pharmaceutical carrier can be liquid, and the pharmaceutical composition can be in the form of a solution.
- Liquid carriers are used to prepare solutions, suspensions, emulsions, syrups, elixirs and pressurized compositions.
- the active ingredients can be dissolved or suspended in a pharmaceutically acceptable liquid carrier, such as water, an organic solvent, a mixture of the two, or a pharmaceutically acceptable oil or fat.
- the pharmaceutical composition for parenteral administration is sterile, substantially isotonic, and pyrogen-free, and is prepared in accordance with the GMP of the FDA or a similar agency.
- the viral vector drug can be administered as an injectable dosage form of a solution or suspension thereof, wherein the substance is in physiologically acceptable diluent and pharmaceutical carrier (which can be a sterile liquid, such as water, oil, saline, glycerol or ethanol).
- physiologically acceptable diluent and pharmaceutical carrier which can be a sterile liquid, such as water, oil, saline, glycerol or ethanol.
- an auxiliary substance such as a wetting agent or an emulsifier, a surfactant and a pH buffering substance can be present in the composition.
- Other components of the pharmaceutical composition include those of petroleum, animal, plant or synthetic origin, such as peanut oil, soybean oil and mineral oil.
- diols such as propylene glycol or polyethylene glycol are preferred liquid carriers, especially for injectable solutions.
- the viral vector drug can be administered in the form of a depot injection or an implanted preparation, which can be formulated to allow sustained release of the active ingredient.
- the composition is prepared as an injectable preparation, i.e., a liquid solution or suspension, or can be prepared as a solid form suitable for dissolution or suspension in a liquid carrier prior to injection.
- nucleotide or “polynucleotide” means a single-stranded or double-stranded deoxyribonucleotide, deoxyribonucleoside, ribonucleoside or ribonucleotide and polymers thereof. Unless specifically limited, the terms cover nucleic acids containing known analogs of natural nucleotides that have binding properties similar to reference nucleic acids and are metabolized in a manner similar to naturally occurring nucleotides. Unless otherwise limited specifically, the terms also mean oligonucleotide analogs, including PNAs (peptide nucleic acids), DNA analogs used in antisense techniques (phosphorothioate, phosphoramidate, etc.), etc.
- PNAs peptide nucleic acids
- DNA analogs used in antisense techniques phosphorothioate, phosphoramidate, etc.
- nucleic acid sequence also implicitly encompasses its conserved modified variants (including, but not limited to, degenerate codon substitutions) and complementary sequences as well as explicitly specified sequences.
- degenerate codon substitution can be achieved by generating a sequence in which the 3rd position of one or more selected (or all) codons is substituted with a mixed base and/or deoxyinosine residue (Batzer et al., Nucleic Acid Res. 19: 5081 (1991); Ohtsuka et al., J. Biol. Chem. 260: 2605-2608 (1985); and Cassol et al., (1992); Rossolini et al., Mol Cell. Probes 8: 91-98 (1994)).
- polypeptide and protein are used interchangeably herein to mean polymers of amino acid residues.
- description of a polypeptide is equally applicable to the description of a peptide and a protein, and vice versa.
- the terms apply to naturally occurring amino acid polymers, and amino acid polymers in which one or more amino acid residues are non-naturally encoded amino acids.
- the terms encompass amino acid chains of any length, including full-length proteins (i.e., antigens), in which amino acid residues are linked via covalent peptide bonds.
- host cell means a cell comprising the nucleotide of the present invention, regardless of the method used for insertion to produce a recombinant host cell, such as direct uptake, transduction, pairing, or other methods known in the art.
- An exogenous polynucleotide can exist as a non-integrated vector such as a plasmid, or can be integrated into the host's genome.
- the host cell can be a prokaryotic cell or a eukaryotic cell.
- transformation means a process by which a heterologous DNA sequence is introduced into a host cell or organism.
- expression means the transcription and/or translation of an endogenous gene or a transgene in a cell.
- the positive and progressive effect of the present invention is as follows:
- the present invention provides a mutant of an immunoglobulin-degrading enzyme, which has higher activity and/or thermal stability than the activity and/or thermal stability of the wild-type IdeE, and higher activity than that of IdeS and IdeZ (which is more effective than IdeS and IdeZ in cleaving human IgG, and the activity of which is nearly twice that of IdeS, and more than 4 times that of IdeZ).
- a mutant library of the wild-type IdeE protein sequence was designed and constructed, and forty mutants were obtained through screening.
- the polynucleotide sequence encoding the wild-type IdeE protein sequence (SEQ ID NO: 2) was synthesized by codon optimization. An N-terminal signal peptide sequence and a C-terminal 6 ⁇ histidine tag were added. A sequence was synthesized, and then inserted into the pET32a expression vector. A recombinant plasmid used to express the wild-type IdeE was obtained after being verified to be correct by sequencing. Based on the expression plasmid of the wild-type IdeE, degenerate primers required for a mutant library were designed. The original wild-type sequence was amplified, and the amplified sequence was inserted into the vector to obtain a mutant library recombinant plasmid.
- the wild-type and mutant library recombinant plasmids were electrotransformed into Escherichia coli BL21 Star (DE3), and the cells were plated on an LB agarose plate containing 100 ug/ml of ampicillin. The plate was cultured at 37° C. overnight until colonies grow out. Single colonies were picked and inoculated to 200 ul of LB medium containing 100 ug/ml of ampicillin, and cultured at 37° C. and 250 rpm overnight. The overnight cultures were inoculated to 1 ml of LB medium containing 100 ug/ml of ampicillin, and cultured at 37° C. for 4 h.
- an ELSIA-based activity assay method was established.
- the principle of the assay was as follows: An ELSIA plate was coated with a human IgG1-specific antigen, and then the supernatant samples containing an equivalent concentration of mutant proteins were incubated with human IgG1 in the wells. A human IgG1 detection antibody specific to the antibody Fc portion was used to measure the amount of intact or partially-cleaved human IgG1 bound to the well.
- IgG1 standard curve can be generated based on the relationship between different concentrations of IgG1 and the corresponding detection signals. According to the standard curve, the amount of intact or partially-cleaved IgG1 was calculated, and then the amount of fully-cleaved IgG1 was calculated. The activity of the mutant was evaluated based on the proportion of fully-cleaved IgG1 to the initial IgG1.
- Example 1 In order that the concentrations of the mutant proteins in the supernatants harvested in Example 1 were equivalent, SDS-PAGE was performed with the same loading amount. The optical density value of a protein band of interest in the electrophoretogram was analyzed using Quantity One. In the case that the loading amounts were the same, the higher the optical density value of the protein band of interest in the pattern was, the higher the concentration was. Using the IdeE supernatant as a control, the other mutant supernatants were concentrated or diluted, so that the optical density values of the mutant protein bands were all substantially the same as that of the IdeE control.
- ELISA was carried out as follows: An ELISA plate was coated with 2 ug/ml of human IgG1 (trastuzumab)-specific antigen (Art. No. QRE-104, RUIAN BIOTECHNOLOGY) at 2-8° C. overnight, and then washed with PBST (PBS+0.05% Tween20). The washed ELISA plate was blocked with 2% BSA (formulated in PBS) at 37° C. for 2 h, and then washed with PBST.
- Cleavage reaction After adjusting the protein concentration, the supernatants were diluted 5 ⁇ with reaction buffer (10 mM PB, 10 mM NaCl, pH 6.5). 50 ul of 100 ng/ml trastuzumab diluent and 50 ul of diluted supernatants were added into the wells of the ELISA plate.
- the ELISA plate was incubated with shaking at 37° C. for 1 h, and washed with PBST. Then, 40 ng/ml of Goat anti-Human IgG Fc Cross-Adsorbed Secondary Antibody-HRP (Art. No. 31413, Thermo) was added into the wells of the plate, and the plate was incubated with shaking at 37° C. for 1 h and washed with PBST. Then, TMB was added as HRP chromogenic substrate. The plate was incubated for 15 min, and then the reaction was terminated with 2N H 2 SO 4 . The absorbance at 450 nm was detected by a microplate reader.
- the concentrations of intact or partially-cleaved trastuzumab in different test wells were calculated, and then the proportions of fully-cleaved trastuzumab to the initial trastuzumab were calculated, so as to evaluate the activity of different mutants.
- the multiple relationship between the activity of each mutant relative to that of the wild-type IdeE is shown in Table 1.
- the forty mutants obtained through screening in Example 1 all had higher activity than that of the wild-type IdeE, of which fifteen mutants were twice or more as active as the wild-type IdeE.
- the supernatant of the wild-type or each mutant was divided into two parts, and the two parts were placed at 4° C. and 50° C. for 1 h, respectively. Subsequently, the activity of the wild-type or each mutant was detected according to the ELISA method in Example 2.
- the residual activity percentage (%) of the wild-type or each mutant after being placed at 50° C. for 1 h was calculated based on the activity after being placed at 50° C./the activity after being placed at 4° C., so as to compare the thermal stability of the wild-type and each mutant.
- the supernatants were further purified with IDA-Ni agarose magnetic beads, and the purified and eluted proteins were buffer exchanged into PBS buffer system with an ultrafiltration centrifuge tube. SDS-PAGE was used to evaluate the purity of the purified mutant proteins. OD280 was detected, and the concentration of the purified mutant proteins was calculated according to the extinction coefficient.
- the activity of cleaving human IgG1 of different mutants relative to the wild-type IdeE was further evaluated via the cleavage products resulting from cleavage of human IgG1 by each mutant at different concentrations displayed on SDS-PAGE.
- the purified mutants and the wild-type IdeE were diluted to 0.002 mg/mL and 0.001 mg/mL, respectively.
- 50 ul of mutants or wild-type IdeE at different concentrations was each added to 50 ul of reaction system containing 2 mg/ml of trastuzumab to initiate the cleavage reaction, and the reaction systems were placed at 37° C. for 30 min.
- the samples were mixed with an equal volume of 2 ⁇ SDS loading buffer, and then placed in water bath at 75° C. for 5 min.
- the cleavage products were detected by SDS-PAGE.
- the first 15 amino acids (D1-V15), the first 16 amino acids (D1-P16), the first 17 amino acids (D1-H17), the first 18 amino acids (D1-Q18) and the first 19 amino acids (D1419) at its N-terminus were respectively deleted, so as to construct five N-terminal truncated mutants (see Table 3).
- Example 2 the polynucleotide sequences of the mutants in Table 3 were synthesized, and the mutant expression recombinant plasmids were constructed to transform Escherichia coli BL21 Star (DE3).
- the mutant purified proteins were prepared according to the method in Example 4.
- the purified mutants and the wild-type IdeE were diluted to 0.002 mg/mL, respectively.
- 50 ul of diluted mutants or wild-type IdeE was each added to 50 ul of reaction system containing 2 mg/ml of trastuzumab to initiate the cleavage reaction, and the reaction systems were placed at 37° C. for 30 min.
- the samples were mixed with an equal volume of 2 ⁇ SDS loading buffer, and then placed in water bath at 75° C. for 5 min.
- the cleavage products were detected by SDS-PAGE.
- the purified mutants and the wild-type IdeE were respectively diluted to 0.1 mg/ml, placed at 50° C. for 1 h, and then further diluted to 0.002 mg/mL.
- 50 ul of diluted mutants or wild-type IdeE was each added to 50 ul of reaction system containing 2 mg/ml of trastuzumab to initiate the cleavage reaction, and the reaction systems were placed at 37° C. for 30 min.
- the samples were mixed with an equal volume of 2 ⁇ SDS loading buffer, and then placed in water bath at 75° C. for 5 min.
- the cleavage products were detected by SDS-PAGE.
- the residual activity of the five truncated mutants after heat treatment at 50° C. was significantly higher than that of the wild-type, indicating that the thermal stability of all of the five truncated mutants is significantly improved compared with that of the wild-type.
- the polynucleotide sequences of the mutants in Table 4 were synthesized, and the mutant expression recombinant plasmids were constructed to transform Escherichia coli BL21 Star (DE3).
- the mutant purified proteins were prepared according to the method in Example 4.
- the purified mutants and the wild-type IdeE were diluted to 0.002 mg/mL, respectively.
- 50 ul of diluted mutants or wild-type IdeE was each added to 50 ul of reaction system containing 2 mg/ml of trastuzumab to initiate the cleavage reaction, and the reaction systems were placed at 37° C. for 30 min.
- the samples were mixed with an equal volume of 2 ⁇ SDS loading buffer, and then placed in water bath at 75° C. for 5 min.
- the cleavage products were detected by SDS-PAGE.
- the cleavage activity of both of the two truncated mutants was more than twice that of the wild-type IdeE.
- Example 2 the polynucleotide sequences of the mutants in Table 5 were synthesized, and the mutant expression recombinant plasmids were constructed to transform Escherichia coli BL21 Star (DE3).
- the mutant purified proteins were prepared according to the method in Example 4.
- the purified mutants were diluted to 0.001 mg/mL, respectively.
- 50 ul of diluted mutants or wild-type IdeE was each added to 50 ul of reaction system containing 2 mg/ml of trastuzumab to initiate the cleavage reaction, and the reaction systems were placed at 37° C. for 30 min.
- the samples were mixed with an equal volume of 2 ⁇ SDS loading buffer, and then placed in water bath at 75° C. for 5 min.
- the cleavage products were detected by SDS-PAGE.
- the purified mutants were respectively diluted to 0.1 mg/ml, placed at 50° C. for 1 h, and then further diluted to 0.001 mg/mL.
- 50 ul of diluted mutants or wild-type IdeE was each added to 50 ul of reaction system containing 2 mg/ml of trastuzumab to initiate the cleavage reaction, and the reaction systems were placed at 37° C. for 30 min.
- the samples were mixed with an equal volume of 2 ⁇ SDS loading buffer, and then placed in water bath at 75° C. for 5 min.
- the cleavage products were detected by SDS-PAGE.
- the purified E97D_del18 mutant in Example 7 was sequentially diluted to 20 ⁇ g/mL, 10 ⁇ g/mL, 5 ⁇ g/mL, 2.5 ⁇ g/mL and 1.25 ⁇ g/ml.
- IdeS FabRICATOR®, Art. No. AO-FRI-020, Genovis
- IdeZ FabRICATOR-Z®, Art. No.
- AO-FRZ-020, Genovis was diluted to 0.4 U/ ⁇ l, 0.2 U/ ⁇ l, 0.1 U/ ⁇ l, 0.05 U/ ⁇ l and 0.025 U/ ⁇ l, respectively.
- 50 ⁇ l of mutants, IdeS or IdeZ at different concentrations was each added to 50 ⁇ l of reaction system containing 2 mg/ml of trastuzumab to initiate the cleavage reaction, and the reaction systems were placed at 37° C. for 30 min.
- the samples were mixed with an equal volume of 2 ⁇ SDS loading buffer, and then placed in water bath at 75° C. for 5 min.
- the cleavage products were detected by SDS-PAGE.
- FIG. 8 shows the electrophoretogram of cleavage products resulting from cleavage of human IgG1 by mutant E97D_del18 and IdeS at different concentrations. From the enzyme protein bands on the electrophoretogram, it can be judged that the concentration of enzyme IdeS in lane 1 was between those of mutant enzyme E97D_del18 in lanes 7 and 8, thus the concentration of enzyme IdeS in lane 3 was deduced to be between those of mutant enzyme E97D_del18 in lanes 9 and 10, and the enzymatic digestion of IgG1 in lane 3 was between those in lanes 10 and 11, and therefore it can be inferred that the activity of cleaving human IgG1 of mutant E97D_del18 is nearly twice that of IdeS.
- FIG. 9 shows the electrophoretogram of cleavage products resulting from cleavage of human IgG1 by mutant E97D_del18 and IdeZ at different concentrations. From the enzyme protein bands on the electrophoretogram, it can be judged that the concentration of enzyme IdeZ in lane 1 was higher than that of mutant enzyme E97D_del18 in lane 7, thus the concentration of enzyme IdeZ in lane 3 was deduced to be higher than that of mutant enzyme E97D_del18 in lane 9, that is, 4 times higher than that of mutant enzyme E97D_del18 in lane 11, and the enzymatic digestion of IgG1 in lane 3 was close to that in lane 11, and therefore it can be inferred that the activity of cleaving human IgG1 of mutant E97D_del18 is 4 times higher than that of IdeZ.
- mice treated with mutant E97D_del18 and human IVIg were evaluated by detecting the amount of intact or single-cleaved IVIg in the sera or plasmas of mice treated with mutant E97D_del18 and human IVIg. According to Table 6, mouse serum or plasma enzymatic digestion systems were formulated for different groups.
- the effect of iodoacetic acid in the iodoacetic acid treatment groups was to inhibit the activity of the IgG-degrading enzyme.
- the systems were placed at 37° C. for 30 min. 20 ⁇ l of samples were mixed with an equal volume of 2 ⁇ SDS non-reducing loading buffer, and then placed in water bath at 75° C. for 5 min. The cleavage products were detected by SDS-PAGE.
- FIG. 10 shows the electrophoretogram of cleavage products resulting from cleavage of human IVIg by mutant E97D_del18 in the sera and plasmas of mice. The results showed that E97D_del18 can effectively cleave human IVIg in both the sera and plasmas of mice.
- mutant E97D_del18 has the activity of cleaving human IgG1 in vitro was evaluated by detecting the sera of mice or humans treated with mutant E97D_del18. According to Table 7, mouse or human serum enzymatic digestion systems were formulated for different groups.
- the systems were placed at 37° C. for 24 h. 20 ⁇ l of samples were mixed with an equal volume of 2 ⁇ SDS reducing loading buffer, then diluted 20 ⁇ with 1 ⁇ SDS reducing loading buffer, and placed in water bath at 75° C. for 5 min. The cleavage products were detected by SDS-PAGE.
- FIG. 11 shows the electrophoretogram of cleavage products produced by mutant E97D_del18 in the sera of mice and humans. The results showed that a significantly visible 25 kD Fc fragment was produced in the sera of humans through E97D_del18 cleavage, which was invisible in the sera of mice, indicating that E97D_del18 can effectively and specifically cleave IgG1 in the sera of humans, and has very low or no activity of cleaving IgG1 in the sera of mice.
- the activity of cleaving serum immunoglobulins of different species of animals in vitro of mutant E97D_del18 was evaluated by detecting the amount of intact or single-cleaved IgG in the sera or plasmas of different species of animals added with mutant E97D_del18. According to Tables 8 and 9, serum or antibody enzymatic digestion systems were formulated for different species.
- Beagle serum and antibody (of different species) enzymatic digestion systems Concentration of Concentration of immunoglobulin E97D_del18 in the system in the system Group name (mg/ml) (mg/ml) Beagle serum control group 10 / Beagle serum enzymatic digestion group 10 0.025/1 Rabbit polyclonal IgG antibody 1 / 1 control group Rabbit polyclonal IgG antibody 1 0.005 1 control group Rabbit polyclonal IgG antibody 1 / 2 control group Rabbit polyclonal IgG antibody 1 0.005 2 control group Mouse monoclonal antibody IgG1 1 0.005 control group Mouse monoclonal antibody IgG1 1 0.005 enzymatic digestion group Mouse monoclonal antibody IgG2a 1 0.005 control group Mouse monoclonal antibody IgG2a 1 0.005 enzymatic digestion group
- the systems were placed at 37° C. for 1 h.
- the enzymatic digestion products were detected by SDS-PAGE.
- FIGS. 12 A- 12 D show the cleavage activity of mutant E97D_del18 on the sera and antibodies of different species.
- the results showed that E97D_del18 can effectively cleave dog IgG, rabbit IgG and mouse IgG2a, but can not cleave mouse IgG1.
- E97D_del18 can effectively cleave IgG in the sera of rabbits, dogs and monkeys, of which IgG in the sera of rabbits was cleaved most effectively, IgG in the sera of pigs was cleaved more effectively, and IgG in the sera of rats and mice was nearly uncleaved.
- the assay was based on the competition between mutant E97D_del18 and IdeS for binding to anti-E97D_del18/IdeS antibodies. Pre-incubation of test enzymes and human serum would enable anti-E97D_del18/IdeS antibodies to bind to mutant E97D_del18 and IdeS.
- a well plate was coated with mutant E97D_del18 and IdeS overnight, then washed with PBST, and blocked in 2% BSA blocking solution for 1 h.
- a mixed plate was prepared with the mutant to be tested and IdeS diluted stepwise and human serum. The mixed plate was incubated with shaking at room temperature for 1 h, and washed with PBST. Then, biotin-labeled E97D_del18 mutant and IdeS were added, followed by SA-HRP. The plate was developed with TMB, and read. Parallel comparison obtained the presence of pre-existing antibodies against E97D_del18 and IdeS in about eighty human blood samples.
- mice Under sterile conditions, two mice were injected with human IVIg (an intravenous human immunoglobulin) at a dose of 1 g/kg intraperitoneally (two mice were parallel experiments, numbered No. 1 and No. 2). 24 h after injecting human IVIg, the mice were injected with the IgG-degrading enzyme (E97D_del18) at a dose of 5 mg/kg intravenously. 0 h, 15 min, 2 h, 6 h and 24 h after injecting E97D_del18, blood was collected from the two mice, and serum samples were collected, respectively.
- human IVIg an intravenous human immunoglobulin
- FIG. 13 shows the electrophoretogram of cleavage products resulting from cleavage of human IVIg by E97D_del18 at different times in the mice. The results showed that E97D_del18 effectively cleaved IVIg in the mice, and the enzymatic digestion was already substantially complete in 15 min.
- Example 1 the polynucleotide sequences of the mutants in Table 11 were synthesized, and the mutant expression recombinant plasmids were constructed to transform Escherichia coli BL21 Star (DE3).
- the mutant purified proteins were prepared according to the method in Example 4.
- the purified mutants were diluted to 0.001 mg/mL, respectively.
- 50 ul of diluted mutants or wild-type IdeE was each added to 50 ul of reaction system containing 2 mg/ml of trastuzumab to initiate the cleavage reaction, and the reaction systems were placed at 37° C. for 30 min.
- the samples were mixed with an equal volume of 2 ⁇ SDS loading buffer, and then placed in water bath at 75° C. for 5 min.
- the cleavage products were detected by SDS-PAGE.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- General Health & Medical Sciences (AREA)
- Microbiology (AREA)
- Zoology (AREA)
- Genetics & Genomics (AREA)
- Organic Chemistry (AREA)
- Wood Science & Technology (AREA)
- Epidemiology (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Immunology (AREA)
- Mycology (AREA)
- Biotechnology (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- Molecular Biology (AREA)
- Biomedical Technology (AREA)
- Endocrinology (AREA)
- Gastroenterology & Hepatology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Virology (AREA)
- Peptides Or Proteins (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
Abstract
Provided is a mutant of an immunoglobulin degrading enzyme IdeE. The immunoglobulin degrading enzyme IdeE includes an amino acid sequence as shown in SEQ ID NO: 2 in the sequence listing. The mutant is obtained by means of replacing at least one or more of the positions 8, 10, 24, 59, 97, and 280 of the amino acid sequence. The function of the mutant includes at least the function of the immunoglobulin degrading enzyme IdeE. The activity and thermal stability of the provided mutant of the immunoglobulin degrading enzyme are higher than those of a wild-type IdeE.
Description
- The present invention relates to the field of biotechnology, in particular to a mutant of an immunoglobulin-degrading enzyme.
- Streptococcus pyogenes is one of the common pathogens in humans and animals. It is widely found in nature and in the oropharyngeal cavity, respiratory tract and intestinal tract of humans or animals. Streptococcal infection may induce related diseases, from milder diseases such as pyogenic dermatitis and pharyngitis, to more serious diseases such as sepsis, necrotizing fasciitis and toxic shock syndrome. Immunoglobulin G-degrading enzyme of Streptococcus pyogenes (IdeS), a common Group A Streptococcus pyogenes (GAS) cysteine protease, has endopeptidase activity that hydrolyzes IgG (Agniswamy J, Lei B, Musser J M et al., J Biol Chem, 2004, 279: 52789-52796. Lei B, DeLeo F R, Reid S D et al., Infect Immun, 2002, 70: 6880-6890. Von Pawel-Rammingen U, Johansson B P, Bjorck L. EMBO J, 2002, 21: 1607-1615.). As a virulence factor of pathogens, it can recognize the CH1 and CH2 domains in the lower hinge regions of antibodies and specifically degrade IgG, to obtain homogeneous F(ab)2 and Fc fragments, helping GASs to evade antibody-mediated phagocytosis and cytotoxicity, thus weakening the killing of GASs by the host's immune system (Von Pawel-Rammingen U. J Innate Immunity, 2012, 4: 132-140. Su, Y.-F. et al., Molecular Immunology, 2011, 49: 134-142.).
- Immunoglobulin G (IgG) is the main antibody component in serum, and accounts for about 75% of serum immunoglobulins. It mainly plays a protective role in the body's immunity and can effectively prevent infectious diseases. In addition to its protective effect, IgG is also related to diseases. In some autoimmune diseases, anti-IgG antibodies react with the body's own molecules, and IgG may cause acute transplant rejection in organ transplantation. IdeS specifically degrades IgG, causing IgG to lose its inherent function, thus achieving immunosuppression.
- At present, IdeS used in clinical practice has the problems of poor activity and high pre-existing antibodies in the human body. IdeS is a virulence factor of human pathogens. Clinical studies have found that anti-IdeS antibodies are detected nearly 100% in normal people under normal physiological conditions, which leads to low administration efficiency and safety concerns in the use of IdeS.
- Protease IdeE with a sequence homology of about 70% to IdeS is derived from a Streptococcus equi subsp. zooepidemicus—Streptococcus equi ssp. equi, an equine pathogen (Jonas Lannergård, Bengt Guss. FEMS Microbiol Lett, 2006, 262: 230-235). The two enzymes, IdeE and IdeS, cleave IgG at exactly the same position. The cleavages are highly reproducible and specific, and have a very similar substrate range. Since IdeE is derived from an equine pathogen, it is speculated that its pre-existing antibodies in the human body may be much lower than IdeS, and it is more suitable for the development of immunosuppressants for treating and preventing diseases mediated by anti-IgG antibodies. However, wild-type IdeE, like IdeS, also has the problem of low activity. Therefore, it is necessary to improve the activity of IdeE through molecular design and mutation screening, reducing doses used in clinical practice, thus reducing the risks posed by high doses of proteins of bacterial origin. This is exactly what the present invention aims to achieve.
- The first aspect of the present invention relates to a mutant of an immunoglobulin-degrading enzyme IdeE, wherein the immunoglobulin-degrading enzyme IdeE comprises or consists of an amino acid sequence as set forth in SEQ ID NO: 2 in the Sequence Listing; and the mutant comprises a mutation selected from the group consisting of:
-
- (1) substitution of one or more of
positions - (2) truncation of the immunoglobulin-degrading enzyme IdeE, by deleting the sequence of the first 1, the first 2, the first 3, the first 4, the first 5, the first 6, the first 7, the first 8, the first 9, the first 10, the first 11, the first 12, the first 13, the first 14, the first 15, the first 16, the first 17, the first 18 or the first 19 amino acids at its N-terminus; and/or
- (3) truncation of the immunoglobulin-degrading enzyme IdeE, by deleting the sequence of the last 1, the last 2, the last 3, the last 4, the last 5, the last 6, the last 7, the last 8, the last 9 or the last 10 amino acids at its C-terminus;
- wherein the mutant has higher activity and/or thermal stability than the activity and/or thermal stability of the immunoglobulin-degrading enzyme IdeE.
- (1) substitution of one or more of
- The second aspect of the present invention relates to a protein comprising the mutant of the present invention. The protein is linked to a secretory signal sequence and/or methionine at the N-terminus of the mutant; and/or the protein is linked to a histidine tag at the C-terminus of the mutant.
- The third to fifth aspects of the present invention relate to a nucleotide encoding the mutant or protein of the present invention, an expression vector comprising the nucleotide, and a host cell comprising the expression vector or expressing the mutant or protein of the present invention.
- The sixth aspect of the present invention relates to a composition or kit, comprising: the mutant or protein of the present invention; and optionally, a substance selected from the group consisting of: a pharmaceutically acceptable carrier or excipient, an antibody or an Fc-containing protein and a viral vector drug.
-
FIG. 1 shows the SDS-PAGE gel electrophoresis pattern of cleavage products resulting from cleavage of human IgG1 by seven single-point mutants and wild-type IdeE (enzyme:substrate=1:1000). -
FIG. 2 shows the SDS-PAGE gel electrophoresis pattern of cleavage products resulting from cleavage of human IgG1 by seven single-point mutants and wild-type IdeE (enzyme:substrate=1:2000). -
FIG. 3 shows the SDS-PAGE gel electrophoresis pattern of cleavage products resulting from cleavage of human IgG1 by five N-terminal truncated mutants (enzyme:substrate=1:1000). -
FIG. 4 shows the SDS-PAGE gel electrophoresis pattern of cleavage products resulting from cleavage of human IgG1 by five N-terminal truncated mutants and wild-type IdeE after being placed at 50° C. for 1 h (enzyme:substrate=1:1000). -
FIG. 5 shows the SDS-PAGE gel electrophoresis pattern of cleavage products resulting from cleavage of human IgG1 by two C-terminal truncated mutants (enzyme:substrate=1:1000). -
FIG. 6 shows the SDS-PAGE gel electrophoresis pattern of cleavage products resulting from cleavage of human IgG1 by five combinatorial mutants (enzyme:substrate=1:2000). -
FIG. 7 shows the SDS-PAGE gel electrophoresis pattern of cleavage products resulting from cleavage of human IgG1 by five combinatorial mutants after being placed at 50° C. for 1 h (enzyme:substrate=1:2000). -
FIG. 8 shows the SDS-PAGE gel electrophoresis pattern of cleavage products resulting from cleavage of human IgG1 by mutant E97D_del18 and IdeS at different concentrations. -
FIG. 9 shows the SDS-PAGE gel electrophoresis pattern of cleavage products resulting from cleavage of human IgG1 by mutant E97D_del18 and IdeZ at different concentrations. -
FIG. 10 shows the SDS-PAGE gel electrophoresis pattern of cleavage products resulting from cleavage of human IVIg by mutant E97D_del18 in the sera and plasmas of mice. -
FIG. 11 shows the SDS-PAGE gel electrophoresis pattern of cleavage products produced by mutant E97D_del18 in the sera of mice and humans. -
FIG. 12A-12D show the SDS-PAGE gel electrophoresis patterns of cleavage products resulting from cleavage of IgG by E97D_del18 at different concentrations in the sera of beagles, rats, mice, rabbits, monkeys and pigs. -
FIG. 13 shows the SDS-PAGE gel electrophoresis pattern of cleavage products resulting from cleavage of human IVIg by E97D_del18 at different times in mice. -
FIG. 14A and 14B show the SDS-PAGE gel electrophoresis patterns of cleavage products resulting from cleavage of human IgG1 by mutants with different mutation combinations and IdeE (enzyme:substrate=1:2000). - I. Functional Polypeptides with Immunoglobulin-Degrading Enzyme Activity
- In the first aspect of the present invention, there is provided a functional polypeptide with the activity of immunoglobulin-degrading enzyme, comprising a mutant based on an amino acid sequence as set forth in SEQ ID NO: 2, wherein the mutant is selected from the group consisting of:
-
- (1) a mutant obtained by substituting one or more amino acids at
positions - (2) an N-terminal truncated mutant of SEQ ID NO: 2, selected from those obtained by deleting the sequence of the first 1, the first 2, the first 3, the first 4, the first 5, the first 6, the first 7, the first 8, the first 9, the first 10, the first 11, the first 12, the first 13, the first 14, the first 15, the first 16, the first 17, the first 18 or the first 19 amino acids at its N-terminus; and/or
- (3) a C-terminal truncated mutant of SEQ ID NO: 2, selected from those obtained by deleting the sequence of the last 1, the last 2, the last 3, the last 4, the last 5, the last 6, the last 7, the last 8, the last 9 or the last 10 amino acids at its C-terminus.
- (1) a mutant obtained by substituting one or more amino acids at
- The mutant of the present invention has the function of the immunoglobulin-degrading enzyme IdeE, and preferably further has improved IgG-cleaving activity and thermal stability.
- The term “having higher activity than that of the immunoglobulin-degrading enzyme IdeE” in the present invention refers to the ability of the mutant to degrade immunoglobulins over that of the wild-type immunoglobulin-degrading enzyme IdeE.
- The term “having higher thermal stability than that of IdeE” in the present invention refers to the ability of the mutant after being maintained at a certain temperature for a period of time to degrade immunoglobulins over that of the wild-type immunoglobulin-degrading enzyme IdeE under the same conditions.
- The mutant of the present invention is preferably produced by means of genetic engineering recombination.
- Preferably, the mutant has at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity to the sequence as set forth in SEQ ID NO: 2.
- More preferably, the amino acid at
position -
- alternatively, the first 15, the first 16, the first 17, the first 18 or the first 19 amino acids are deleted at the N-terminus of the immunoglobulin-degrading enzyme IdeE, for example, the amino acid sequence of the resulting mutant is as set forth in any one of SEQ ID NOs: 18-22;
- alternatively, the last 1, the last 5, the last 8 or the last 10 amino acids are deleted at the C-terminus of the immunoglobulin-degrading enzyme IdeE, for example, the amino acid sequence of the resulting mutant is as set forth in any one of SEQ ID NOs: 23-24;
- alternatively, the amino acid at
position - alternatively, the amino acid at
position
- In a preferred embodiment of the present invention, the amino acid substitution is selected from the group consisting of:
-
- (1) the threonine at
position 8 of SEQ ID NO: 2 is substituted with any one of cysteine, phenylalanine, tryptophan, tyrosine, aspartic acid, glutamic acid, alanine, glycine, histidine, isoleucine, leucine, methionine, asparagine, proline, glutamine, serine, valine, arginine and lysine; - (2) the alanine at
position 10 of SEQ ID NO: 2 is substituted with any one of cysteine, aspartic acid, glutamic acid, phenylalanine, glycine, histidine, isoleucine, lysine, leucine, methionine, asparagine, proline, glutamine, arginine, serine, threonine, valine, tryptophan and tyrosine; - (3) the threonine at position 24 of SEQ ID NO: 2 is substituted with any one of alanine, cysteine, aspartic acid, asparagine, glutamic acid, phenylalanine, glycine, histidine, isoleucine, lysine, leucine, methionine, proline, glutamine, arginine, serine, valine, tryptophan and tyrosine;
- (4) the alanine at position 59 of SEQ ID NO: 2 is substituted with any one of cysteine, aspartic acid, glutamic acid, phenylalanine, glycine, histidine, isoleucine, lysine, leucine, methionine, asparagine, proline, glutamine, arginine, serine, threonine, valine, tryptophan and tyrosine;
- (5) the glutamic acid at position 97 of SEQ ID NO: 2 is substituted with any one of alanine, cysteine, aspartic acid, phenylalanine, glycine, histidine, isoleucine, lysine, leucine, methionine, asparagine, proline, glutamine, arginine, serine, threonine, valine, tryptophan and tyrosine; and
- (6) the arginine at position 280 of SEQ ID NO: 2 is substituted with any one of alanine, aspartic acid, glutamic acid, cysteine, serine, phenylalanine, histidine, isoleucine, lysine, leucine, methionine, asparagine, proline, glutamine, threonine, valine, tryptophan and tyrosine.
- (1) the threonine at
- In a more preferred embodiment of the present invention, the amino acid substitution is selected from the group consisting of:
-
- (1) the threonine at
position 8 of SEQ ID NO: 2 is substituted with aspartic acid, glutamic acid, tryptophan or tyrosine; - (2) the alanine at
position 10 of SEQ ID NO: 2 is substituted with lysine or arginine; - (3) the threonine at position 24 of SEQ ID NO: 2 is substituted with alanine, glycine or serine;
- (4) the alanine at position 59 of SEQ ID NO: 2 is substituted with isoleucine, leucine or valine;
- (5) the glutamic acid at position 97 of SEQ ID NO: 2 is substituted with asparagine; and/or
- (6) the arginine at position 280 of SEQ ID NO: 2 is substituted with histidine or lysine.
- (1) the threonine at
- In another preferred embodiment, based on the five sequences of SEQ ID NO: 9, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 1, SEQ ID NO: 5 and SEQ ID NO: 16 obtained through amino acid substitution, the first 18 amino acids at their N-termini are further deleted, and the amino acid sequences of the resulting mutants are as set forth in SEQ ID NOs: 25-29 in the Sequence Listing.
- In another preferred embodiment, based on the five sequences of SEQ ID NOs: 26-29 obtained through amino acid substitution, the 5 or 10 amino acids at their C-termini are further deleted, and the amino acid sequences of the resulting mutants are as set forth in SEQ ID NOs: 30-34 in the Sequence Listing.
- In another preferred embodiment, the combinatorial mutation is further performed based on the three mutants of SEQ ID NOs:14-16, and the amino acid sequence of the resulting mutant is as set forth in SEQ ID NO: 35 in the Sequence Listing. In another preferred embodiment, based on the five sequences of SEQ ID NO: 9, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 1, SEQ ID NO: 5 and SEQ ID NO: 16 obtained through amino acid substitution, the first 18 amino acids at their N-termini are further deleted, and the amino acid sequences of the resulting mutants are as set forth in SEQ ID NOs: 25-29 in the Sequence Listing.
- Preferably, the mutant of the present invention can also be further mutated, and the sequence of the variant obtained through further mutation has at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity to the sequence of SEQ ID NO: 2, and the variant also has the function of the immunoglobulin-degrading enzyme IdeE.
- The complete sequence of the IdeE used in the present invention is publicly available under the GenBank accession number ABF57910.1, and its sequence is provided herein as SEQ ID NO: 1. The sequence comprises an N-terminal methionine, followed by a 33-amino acid secretory signal sequence, followed by an IdeE coding sequence. The N-terminal methionine and the signal sequence are usually removed to form a mature IdeE protein, the sequence of which is provided herein as SEQ ID NO: 2. Unless otherwise stated, all references to the numbers of the amino acid positions in the sequences of the immunoglobulin-degrading enzymes disclosed herein are based on the numbers of the corresponding positions in SEQ ID NO: 2 starting from the N-terminus.
- The present invention also provides a protein comprising the mutant as described above.
- In a preferred embodiment, the protein comprises a signal peptide at the N-terminus of the mutant described above; preferably, the protein is linked to a secretory signal sequence at the N-terminus of the mutant, and to methionine at the N-terminus of the secretory sequence, and/or is linked to a histidine tag at the C-terminus of the mutant; more preferably, the protein comprises or consists of the following from the N-terminus to the C-terminus: methionine, the secretory signal sequence and the mutant.
- In the second aspect of the present invention, there is provided a nucleotide encoding the protein or mutant as described above.
- The present invention also provides an expression vector comprising the nucleotide.
- The present invention also provides a host cell comprising the expression vector as described above, or a host cell expressing the protein or mutant as described above.
- The host cell can be a conventional cell for expressing proteins or polypeptides in the art, and is selected from E. coli cells, yeast cells, etc.
- II. Pharmaceutical Combinations
- In the third aspect of the present invention, there is provided a composition, comprising an immunoglobulin-degrading enzyme or a mutant thereof or a protein comprising an immunoglobulin-degrading enzyme or a mutant thereof; and optionally, a pharmaceutically acceptable carrier or excipient. In a specific embodiment, the immunoglobulin-degrading enzyme is selected from IdeE, IdeS and IdeZ. In a specific embodiment, the mutant of the immunoglobulin-degrading enzyme is a mutant as described above, and the protein is a protein comprising the mutant as described above. In a specific embodiment, the composition of the present invention further comprises: an antibody or an Fc-containing protein. In a specific embodiment, the antibody target is selected from the group consisting of: a cell surface protein, a cytokine, a hormone, an enzyme, an intracellular messenger, an intercellular messenger and an immune checkpoint. In a specific embodiment, the composition of the present invention further comprises: a viral vector drug, preferably selected from the group consisting of: an oncolytic virus, a gene therapy virus and a viral vector vaccine. In a specific embodiment, the composition of the present invention further comprises: an agent capable of reducing the IgG level in the blood, preferably selected from the group consisting of: an FcRn antibody and an Fc fragment variant with a high affinity to FcRn.
- 2.1. Antibody Targets
- Preferably, in the composition as described above, the antibody target can be a cell surface protein, including but not limited: AFP, αv integrin, α4β7 integrin, BCMA, CD2, CD3, CD19, CD20, CD22, CD25, CD30, CD32, CD33, CD36, CD40, CD46, CD52, CD56, CD64, CD70, CD74, CD79, CD80, CD86, CD105, CD121, CD123, CD133, CD138, CD174, CD205, CD227, CD326, CD340, CEA, c-Met, Cripto, CA1X, Claudin18.2, ED-B, EGFR, EpCAM, EphA2, EphB2, FAP, FOLR1, GD2, Globo H, GPC3, GPNMB, HER-1, HER-2, HER-3, MAGE-A3, mesothelin, MUC16, GPNMB, PSMA, TMEFF2, TAG-72, 5T4, ROR-1, Sca-1, SP, VEGF or WT1.
- The antibody target can be a cytokine, including but not limited to: interleukins IL-1 to IL-13, tumor necrosis factors α and β, interferons α, β and γ, tumor growth factor β (TGF-β) colony stimulating factor (CSF) or granulocyte-monocyte colony stimulating factor (GM-CSF). See Human Cytokines: Handbook for Basic & Clinical Research (Aggrawal et al. (eds), Blackwell Scientific, Boston, MA 1991).
- The antibody target can be a hormone, an enzyme, and an intracellular and intercellular messenger, such as adenylate cyclase, guanylate cyclase or phospholipase C.
- The antibody target can be an immune checkpoint, including: CTLA-4, PD-1, PD-L1, TIM-3, LAG3, Siglec15, 4-1BB, GITR, OX40, CD40L, CD28, TIGIT and VISTA.
- 2.2. Targeted Drugs
- Preferably, in the composition as described above, the combination further comprises a targeted drug or a chemotherapeutic drug or an immune checkpoint blocker, wherein the targeted drug is selected from an epigenetic drug, such as a histone deacetylase inhibitor, an inhibitor targeting the PI3K/Akt/mTOR signaling pathway, such as Tricibine, and a tyrosine kinase inhibitor, such as sunitinib; the chemotherapeutic drug is selected from an immunosuppressant, such as cyclophosphamide, thalidomide and pomalidomide, a proteasome inhibitor, such as bortezomib, a cytotoxic drug, such as gemcitabine and temozolomide, and a cell cycle non-specific drug, such as mitoxantrone; and the immune checkpoint blocker is selected from an anti-CTLA-4 antibody, an anti-PD-1 antibody, an anti-TIM-3 antibody, an anti-LAG3 antibody, an anti-Siglec15 antibody, an anti-4-1BB antibody, an anti-GITR antibody, an anti-OX40 antibody, an anti-CD40L antibody, an anti-CD28 antibody, an anti-TIGIT antibody and an anti-VISTA antibody.
- 2.3. Agents Capable of Reducing Blood IgG Level
- Preferably, in the composition as described above, the polypeptide drug capable of reducing the IgG level in the blood can block the binding of IgG in the blood to the FcRn protein. Preferably, the affinity of the polypeptide to human FcRn protein is higher than that of IgG in the blood to human FcRn protein; and the IgG is selected from IgG1, IgG2, IgG3 and IgG4. Preferably, the polypeptide comprises an antibody Fc fragment variant, which comprises a mutation capable of increasing the affinity of Fc to FcRn, preferably YTE, YTEKF, LS or NHS, wherein the antibody Fc fragment is, for example, Efgartigimod. The variant can be a monomer, a dimer or a multimer. The positions of the mutations of YTE, YTEKF, LS, NHS, etc., which can be used in the present invention, are as described by Dall'Acqua et al. (WF, D. A. et al. (2002). Journal of immunology (Baltimore, Md.: 1950) 169(9): 5171-5180.) and Lee et al. (Lee, C. H. et al. (2019). Nat Commun 10(1): 5031.), respectively. The mutation is performed on one selected from human IgG, which is selected from IgG1, IgG2, IgG3 and IgG4.
- Other Fc fragment variants, which can be used in the present invention, comprise mutations including but not limited to those as described by Dall'Acqua et al. (WF, D. A. et al. (2002). Journal of immunology (Baltimore, Md.: 1950) 169(9): 5171-5180.), Shan et al. (Shan, L. et al. (2016). PLoS One 11(8): e0160345.), Lee et al. (Lee, C. H. et al. (2019). Nat Commun 10(1): 5031.), Mackness et al. (Mackness, B. C. et al. (2019). MAbs 11(7): 1276-1288.) and Christophe et al. (Dumet Christophe, Pottier Jérémy, Gouilleux-Gruart Valérie et al., MAbs, 2019, 11: 1341-1350.).
- Preferably, the polypeptide comprises an antibody Fc fragment variant, which comprises a mutation capable of increasing the affinity of Fc to FcγR, preferably S239D/I322E, S239D/I322E/A330L, K326W/E333S or R214K mutation, preferably afucosylation modification. The variant can be a monomer, a dimer or a multimer. Other Fc fragment variants, which can be used in the present invention, comprise mutations including but not limited to those as described by Wang et al. (Wang Xinhua., Mathieu Mary., Brezski Randall J. (2018). Protein Cell, 9(1), 63-73. doi: 10.1007/s13238-017-0473-8).
- Preferably, the variant comprising a mutation capable of increasing the affinity of Fc to FcRn also comprises a mutation capable of increasing the affinity of Fc to FcγR. The variant can be a monomer, a dimer or a multimer.
- Preferably, in the pharmaceutical combination as described above, the polypeptide is selected from an anti-FcRn antibody, such as Nipocalimab, Rozanolixizumab, RVT-1401, HBM9161, ALXN1830, SYNT001 or Nirsevimab.
- Preferably, in the pharmaceutical combination as described above, the polypeptide is selected from a small peptide fragment capable of specifically binding to FcRn and with a length of 10-70 amino acids, such as ABY-039.
- Preferably, the polypeptide is selected from an Fc multimer capable of specifically binding to FcRn, such as GL-2045, M230, PRIM, HexaGard™, CSL777 or hexavalent molecules by UCB.
- Preferably, the polypeptide includes, but is not limited to, a polypeptide fragment as described by Sockolosky et al. (Sockolosky Jonathan T, Szoka Francis C. Adv. Drug Deliv. Rev., 2015, 91: 109-24).
- 2.4. Viral Vector Drugs
- Preferably, in the viral vector drug of the composition as described above, the virus used in the viral vector drug is selected from an ssDNA virus, a dsDNA virus, an ssRNA virus or a dsRNA virus; and/or the virus used in the viral vector drug is selected from a wild-type virus strain or naturally attenuated strain, a genetically engineered and selective attenuated strain, a gene-loaded virus strain and a gene transcription-targeting virus strain.
- Preferably, the wild-type virus strain or naturally attenuated strain is selected from Newcastle disease virus, reovirus, mumps virus, West Nile virus, adenovirus, vaccinia virus, etc.
- Preferably, the genetically engineered and selective attenuated strain enables the virus to selectively replicate in a tumor by manually deleting a key gene, such as thymidine kinase (TK)-knockout genetically modified human herpes simplex virus I (HSV-1), and the genetically engineered and selective attenuated strain is, for example, ONYX-015 or G207. In ONYX-015, a segment of 827 bp in the E1b region is deleted, and a point mutation is made at the gene for protein E1B55K, so that its expression is terminated prematurely, and the E1B55K protein can not be expressed. In G207, the γ34.5 gene is deleted, which is the determinant of HSV-1 neurotoxicity.
- Preferably, the gene-loaded virus strain is loaded with an exogenous gene, such as one of granulocyte-macrophage colony stimulating factor (GM-CSF), and the gene-loaded virus strain is, for example, JX-594 or T-VEC.
- Preferably, the gene transcription-targeting virus strain enables to control the replication of the oncolytic virus in a tumor cell by inserting a tissue- or tumor-specific promoter upstream of an essential gene of the virus, and the gene transcription-targeting virus strain is, for example, G92A.
- Preferably, in the pharmaceutical combination as described above, the ssDNA virus is selected from parvovirus, preferably H-1PV virus.
- Preferably, the dsDNA virus is selected from herpes simplex virus, adenovirus and poxvirus; more preferably, the herpes simplex virus is preferably herpes simplex virus type I (HSV-1), such as R3616, T-VEC, HF10, G207, NV1020 and OrienX010; the poxvirus is selected from Pexa-Vec (a vaccinia virus), JX-594 (a vaccinia virus), GL-ONC1 and Myxoma; and the adenovirus is selected from Enadenotucirev, DNX-2401, C-REV, NG-348, ProsAtak, CG0070, ADV-TK, EDS01, KH901, H101, H103, VCN-01 and Telomelysin (OBP-301).
- Preferably, the ssRNA virus is selected from picornavirus, alphavirus, retrovirus, paramyxovirus and rhabdovirus; preferably, the picornavirus is selected from CAVATAK, PVS-RIPO, CVA21 (an enterovirus) and RIGVIR, the alphavirus is selected from M1, Sindbis AR339 and Semliki Forest virus, the retrovirus is selected from Toca511, the paramyxovirus is selected from MV-NIS and PV701 (a Newcastle disease virus), and the rhabdovirus is selected from VSV-IFNβ, MG1-MAGEA3 and VSV-GP.
- Preferably, the dsRNA virus is selected from reovirus; preferably, the reovirus is selected from Pelareorep, Reolysin, vaccinia virus, mumps virus and human immunodeficiency virus (HIV). Preferably, the RNA virus is selected from reovirus, coxsackievirus, poliovirus, porcine Seneca Valley virus, measles virus, Newcastle disease virus, vesicular stomatitis virus and influenza virus.
- Preferably, in the pharmaceutical combination as described above, the oncolytic virus expresses an exogenous gene, preferably those of a Bispecific T cell engager (BiTE), an scFv fragment, a cytokine and a chemokine. The BiTE can bind to a molecule that activates T cells such as
CD 3, and can also bind to an antigen target on the surface of a cancer cell. The scFv targets an immune checkpoint, including CTLA-4, PD-1, TIM-3, LAG3, Siglec15, 4-1BB, GITR, OX40, CD40L, CD28, TIGIT and VISTA. The cytokine and chemokine are, for example, GM-CSF, interleukin-2 (IL-2), interleukin-12 (IL-12), an interferon (IFN), a tumor necrosis factor (TNF), soluble CD80 and CCL3. - 2.5. Gene Therapy Drugs
- Preferably, in the composition as described above, the gene therapy virus expresses an exogenous gene, which encodes a protein required for a gene-deficient disease selected from acidic α-glucosidase, copper-transporting ATPase 2, α-galactosidase, argininosuccinate synthase, β-glucocerebrosidase, β-hexosaminidase A, Cl protease inhibitors or Cl esterase inhibitors, glucose 6-phosphatase, insulin, glucagon, growth hormone, parathyroid hormone, growth hormone releasing factor, follicle stimulating hormone, luteinizing hormone, human chorionic gonadotropin, vascular endothelial growth factor, angiopoietin, angiostatin, granulocyte colony-stimulating factor, erythropoietin, connective tissue growth factor, basic fibroblast growth factor, acidic fibroblast growth factor, epidermal growth factor, transforming growth factor α, platelet-derived growth factor, insulin growth factors I and II, TGF, bone morphogenetic protein, nerve growth factor, brain-derived neurotrophic factor, neurotrophins NT-3 and NT4/5, ciliary neurotrophic factor, glial cell line-derived neurotrophic factor, neurotrophin, lectin, netrin-1 and netrin-2, hepatocyte growth factor, ephrins, tyrosine hydroxylase, thrombopoietin, interleukins (IL-1 to IL-36, etc.), monocyte chemoattractant protein, leukemia inhibitory factor, granulocyte-macrophage colony-stimulating factor, Fas ligand, tumor necrosis factors a and b, interferon a/b/g, stem cell factor, flk-2/flt3 ligand, IgM, IgA, IgD and IgE, chimeric immunoglobulins, humanized antibodies, single-chain antibodies, T cell receptors, chimeric T cell receptors, single-chain T cell receptors, MHC class I and class II molecules, cystic fibrosis transmembrane regulatory factor, coagulation (clotting) factors (factor XIII, factor IX, factor VIII, factor X, factor VII, factor VIIa, protein C, etc.), retinal pigment epithelium-specific 65 kDa protein, LDL receptor, lipoprotein lipase, ornithine transcarbamylase, β-globulin, α-globulin, spectrin, α-antitrypsin, adenosine deaminase, metal transporter (ATP7A or ATP7), sulfonamidase, enzymes involved in lysosomal storage disease (ARSA), hypoxanthine-guanine phosphoribosyltransferase, b-25 glucocerebrosidase, sphingomyelinase, lysosomal hexosaminidase and branched-chain keto acid dehydrogenase.
- Preferably, in the composition as described above, the gene therapy virus carries an exogenous gene, which encodes an inhibitory nucleic acid selected from siRNA, antisense molecules, miRNA, RNAi, ribozymes and shRNA. The inhibitory nucleic acid binds to a polynucleotide repeat disease-related gene, the transcript of which or the polynucleotide of the transcript of which is repeated. The disease-related gene encodes a related protein selected from huntington protein (HTT), androgen receptor on the X chromosome in spinobulbar muscular atrophy, human Ataxin-1/-2/-3/-7, Cav2.1 P/Q voltage-dependent calcium channel (CACNA1A), TATA-binding protein, Ataxin8 opposite strand (ATXN80S), serine/threonine protein phosphatase 2A 55 kDa regulatory subunit B beta isoform in spinocerebellar ataxia (
types FMR2 family member 2 in fragile XE mental retardation, myotonin-protein kinase (MT-PK) in myotonic dystrophy and Frataxin. The disease-related gene is selected from a mutant of superoxide dismutase 1 (SOD1) gene, a gene related to pathogenesis of Parkinson's disease and/or Alzheimer's disease, apolipoprotein B (APOB) gene, PCSK9 gene, HIV infection-related genes (HIVTat, TAR, HIVTAR and CCR5), influenza A virus genome/gene sequences in influenza virus infection, severe acute respiratory syndrome (SARS) coronavirus genome/gene sequences in SARS infection, respiratory syncytial virus genome/gene sequences in respiratory syncytial virus infection, Ebola virus genome/gene sequences in Ebola virus infection, hepatitis B and C virus genome/gene sequences in hepatitis B and C virus infection, herpes simplex virus (HSV) genome/gene sequences in HSV infection, coxsackievirus B3 genome/gene sequences in coxsackievirus B3 infection, silencing of a pathogenic allele of a gene (allele-specific silencing) like torsinA in primary dystonia, pan-class I and HLA-allele specific in transplant, and mutant rhodopsin gene in autosomal dominantly inherited retinitis pigmentosa. - III. Articles of Manufacture
- The present invention also provides an article of manufacture, comprising the mutant or protein as described above; and a therapeutic agent selected from a viral vector drug, an antibody, and a polypeptide drug capable of reducing the IgG level in the blood.
- The present invention also provides a kit or kit of parts, comprising: 1) a therapeutically effective amount of an agent, including the mutant as described above; and 2) a therapeutically effective amount of a therapeutic agent selected from a viral vector drug, an antibody, and a polypeptide drug capable of reducing the IgG level in the blood; preferably, the viral vector drug is an oncolytic virus or a gene therapy virus. The kit can further comprise 3) a targeted drug or a chemotherapeutic drug or an immune checkpoint blocker. The targeted drug is selected from an epigenetic drug, such as a histone deacetylase inhibitor, an inhibitor targeting the PI3K/Akt/mTOR signaling pathway, such as Tricibine, and a tyrosine kinase inhibitor, such as sunitinib; the chemotherapeutic drug is selected from an immunosuppressant, such as cyclophosphamide, thalidomide and pomalidomide, a proteasome inhibitor, such as bortezomib, a cytotoxic drug, such as gemcitabine and temozolomide, and a cell cycle non-specific drug, such as mitoxantrone; and the immune checkpoint blocker is selected from an anti-CTLA-4 antibody, an anti-PD-1 antibody, an anti-TIM-3 antibody, an anti-LAG3 antibody, an anti-Siglec15 antibody, an anti-4-1BB antibody, an anti-GITR antibody, an anti-OX40 antibody, an anti-CD40L antibody, an anti-CD28 antibody, an anti-TIGIT antibody and an anti-VISTA antibody.
- The kit or kit of parts comprises a part A, comprising a therapeutically effective amount of the mutant or protein as described above; and a part B, comprising a therapeutically effective amount of a therapeutic agent selected from a viral vector drug, preferably an oncolytic virus or a gene therapy virus, an antibody, and a polypeptide drug capable of reducing the IgG level in the blood. The kit of parts can further comprise a part C. The part C comprises a targeted drug or a chemotherapeutic drug or an immune checkpoint blocker. The targeted drug is selected from an epigenetic drug, such as a histone deacetylase inhibitor, an inhibitor targeting the PI3K/Akt/mTOR signaling pathway, such as Tricibine, and a tyrosine kinase inhibitor, such as sunitinib; the chemotherapeutic drug is selected from an immunosuppressant, such as cyclophosphamide, thalidomide and pomalidomide, a proteasome inhibitor, such as bortezomib, a cytotoxic drug, such as gemcitabine and temozolomide, and a cell cycle non-specific drug, such as mitoxantrone; and the immune checkpoint blocker is selected from an anti-CTLA-4 antibody, an anti-PD-1 antibody, an anti-TIM-3 antibody, an anti-LAG3 antibody, an anti-Siglec15 antibody, an anti-4-1BB antibody, an anti-GITR antibody, an anti-OX40 antibody, an anti-CD40L antibody, an anti-CD28 antibody, an anti-TIGIT antibody and an anti-VISTA antibody.
- The kit can comprise instructions on the administration of the therapeutically effective amount of the mutant or protein as described above and the therapeutically effective amount of the therapeutic agent (e.g., dose information and administration interval information). The therapeutic agent is selected from a viral vector drug, preferably an oncolytic virus or a gene therapy virus, an antibody, and a polypeptide drug capable of reducing the IgG level in the blood.
- Well-established expression systems can be used to prepare viral vector drugs. Some examples of methods include the use of mammalian cell expression systems to produce viral particles, such as the use of HEK293 cells to produce adenovirus viral vector drugs (Freedman Joshua D, Duffy Margaret R, Lei-Rossmann Janet et al., An Oncolytic Virus Expressing a T-cell Engager Simultaneously Targets Cancer and Immunosuppressive Stromal Cells. [J]. Cancer Res., 2018, 78: 6852-6865).
- The pharmaceutical carrier can be liquid, and the pharmaceutical composition can be in the form of a solution. Liquid carriers are used to prepare solutions, suspensions, emulsions, syrups, elixirs and pressurized compositions. The active ingredients can be dissolved or suspended in a pharmaceutically acceptable liquid carrier, such as water, an organic solvent, a mixture of the two, or a pharmaceutically acceptable oil or fat.
- The pharmaceutical composition for parenteral administration is sterile, substantially isotonic, and pyrogen-free, and is prepared in accordance with the GMP of the FDA or a similar agency. The viral vector drug can be administered as an injectable dosage form of a solution or suspension thereof, wherein the substance is in physiologically acceptable diluent and pharmaceutical carrier (which can be a sterile liquid, such as water, oil, saline, glycerol or ethanol). In addition, an auxiliary substance such as a wetting agent or an emulsifier, a surfactant and a pH buffering substance can be present in the composition. Other components of the pharmaceutical composition include those of petroleum, animal, plant or synthetic origin, such as peanut oil, soybean oil and mineral oil. In general, diols such as propylene glycol or polyethylene glycol are preferred liquid carriers, especially for injectable solutions. The viral vector drug can be administered in the form of a depot injection or an implanted preparation, which can be formulated to allow sustained release of the active ingredient. Typically, the composition is prepared as an injectable preparation, i.e., a liquid solution or suspension, or can be prepared as a solid form suitable for dissolution or suspension in a liquid carrier prior to injection.
- Unless otherwise defined, all technical and scientific terms used herein have the same meanings as commonly understood by those of ordinary skill in the art to which the present invention belongs. While any methods, devices and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, preferred methods, devices and materials are now described.
- The term “nucleotide” or “polynucleotide” means a single-stranded or double-stranded deoxyribonucleotide, deoxyribonucleoside, ribonucleoside or ribonucleotide and polymers thereof. Unless specifically limited, the terms cover nucleic acids containing known analogs of natural nucleotides that have binding properties similar to reference nucleic acids and are metabolized in a manner similar to naturally occurring nucleotides. Unless otherwise limited specifically, the terms also mean oligonucleotide analogs, including PNAs (peptide nucleic acids), DNA analogs used in antisense techniques (phosphorothioate, phosphoramidate, etc.), etc. Unless otherwise specified, a particular nucleic acid sequence also implicitly encompasses its conserved modified variants (including, but not limited to, degenerate codon substitutions) and complementary sequences as well as explicitly specified sequences. In particular, degenerate codon substitution can be achieved by generating a sequence in which the 3rd position of one or more selected (or all) codons is substituted with a mixed base and/or deoxyinosine residue (Batzer et al., Nucleic Acid Res. 19: 5081 (1991); Ohtsuka et al., J. Biol. Chem. 260: 2605-2608 (1985); and Cassol et al., (1992); Rossolini et al., Mol Cell. Probes 8: 91-98 (1994)).
- The terms “polypeptide” and “protein” are used interchangeably herein to mean polymers of amino acid residues. In other words, the description of a polypeptide is equally applicable to the description of a peptide and a protein, and vice versa. The terms apply to naturally occurring amino acid polymers, and amino acid polymers in which one or more amino acid residues are non-naturally encoded amino acids. As used herein, the terms encompass amino acid chains of any length, including full-length proteins (i.e., antigens), in which amino acid residues are linked via covalent peptide bonds.
- The term “host cell” means a cell comprising the nucleotide of the present invention, regardless of the method used for insertion to produce a recombinant host cell, such as direct uptake, transduction, pairing, or other methods known in the art. An exogenous polynucleotide can exist as a non-integrated vector such as a plasmid, or can be integrated into the host's genome. The host cell can be a prokaryotic cell or a eukaryotic cell.
- The term “transformation” means a process by which a heterologous DNA sequence is introduced into a host cell or organism.
- The term “expression” means the transcription and/or translation of an endogenous gene or a transgene in a cell.
- The positive and progressive effect of the present invention is as follows: The present invention provides a mutant of an immunoglobulin-degrading enzyme, which has higher activity and/or thermal stability than the activity and/or thermal stability of the wild-type IdeE, and higher activity than that of IdeS and IdeZ (which is more effective than IdeS and IdeZ in cleaving human IgG, and the activity of which is nearly twice that of IdeS, and more than 4 times that of IdeZ).
- The present invention will be further described by way of examples below, but the present invention is not limited to the scope of the described examples. The experimental methods in the following examples that do not indicate the specific conditions are selected according to conventional methods and conditions or according to the product's instructions.
- A mutant library of the wild-type IdeE protein sequence was designed and constructed, and forty mutants were obtained through screening.
- The polynucleotide sequence encoding the wild-type IdeE protein sequence (SEQ ID NO: 2) was synthesized by codon optimization. An N-terminal signal peptide sequence and a C-terminal 6×histidine tag were added. A sequence was synthesized, and then inserted into the pET32a expression vector. A recombinant plasmid used to express the wild-type IdeE was obtained after being verified to be correct by sequencing. Based on the expression plasmid of the wild-type IdeE, degenerate primers required for a mutant library were designed. The original wild-type sequence was amplified, and the amplified sequence was inserted into the vector to obtain a mutant library recombinant plasmid. The wild-type and mutant library recombinant plasmids were electrotransformed into Escherichia coli BL21 Star (DE3), and the cells were plated on an LB agarose plate containing 100 ug/ml of ampicillin. The plate was cultured at 37° C. overnight until colonies grow out. Single colonies were picked and inoculated to 200 ul of LB medium containing 100 ug/ml of ampicillin, and cultured at 37° C. and 250 rpm overnight. The overnight cultures were inoculated to 1 ml of LB medium containing 100 ug/ml of ampicillin, and cultured at 37° C. for 4 h. Then, 0.1 mM of IPTG was added, and the mixtures were further cultured at 30° C. overnight. Supernatants were harvested by centrifugation from the overnight cultures. SDS-PAGE was used to evaluate the concentrations of the mutant proteins in the mutant expression supernatants.
- In order to evaluate the activity of cleaving human IgG1 of each mutant, an ELSIA-based activity assay method was established. The principle of the assay was as follows: An ELSIA plate was coated with a human IgG1-specific antigen, and then the supernatant samples containing an equivalent concentration of mutant proteins were incubated with human IgG1 in the wells. A human IgG1 detection antibody specific to the antibody Fc portion was used to measure the amount of intact or partially-cleaved human IgG1 bound to the well. In the case that the concentrations of the mutant proteins in the given supernatants in the wells were the same, the higher the activity of cleaving human IgG1 of the mutant protein was, the less intact anti-human IgG1 antibody bound to the well, thus obtaining a lower signal. An IgG1 standard curve can be generated based on the relationship between different concentrations of IgG1 and the corresponding detection signals. According to the standard curve, the amount of intact or partially-cleaved IgG1 was calculated, and then the amount of fully-cleaved IgG1 was calculated. The activity of the mutant was evaluated based on the proportion of fully-cleaved IgG1 to the initial IgG1.
- In order that the concentrations of the mutant proteins in the supernatants harvested in Example 1 were equivalent, SDS-PAGE was performed with the same loading amount. The optical density value of a protein band of interest in the electrophoretogram was analyzed using Quantity One. In the case that the loading amounts were the same, the higher the optical density value of the protein band of interest in the pattern was, the higher the concentration was. Using the IdeE supernatant as a control, the other mutant supernatants were concentrated or diluted, so that the optical density values of the mutant protein bands were all substantially the same as that of the IdeE control.
- After adjusting the protein concentrations in the supernatants to an equivalent level, ELISA was carried out as follows: An ELISA plate was coated with 2 ug/ml of human IgG1 (trastuzumab)-specific antigen (Art. No. QRE-104, RUIAN BIOTECHNOLOGY) at 2-8° C. overnight, and then washed with PBST (PBS+0.05% Tween20). The washed ELISA plate was blocked with 2% BSA (formulated in PBS) at 37° C. for 2 h, and then washed with PBST.
- Generation of standard curve: 200 ng/ml of trastuzumab solution was subjected to gradient dilution with reaction buffer (10 mM PB, 10 mM NaCl, pH 6.5) at a ratio of 1:2 to 3.125 ng/ml. 100 ul of diluents with different concentrations of trastuzumab were added into the wells of the ELISA plate for the generation of a standard curve of substrate (trastuzumab).
- Cleavage reaction: After adjusting the protein concentration, the supernatants were diluted 5×with reaction buffer (10 mM PB, 10 mM NaCl, pH 6.5). 50 ul of 100 ng/ml trastuzumab diluent and 50 ul of diluted supernatants were added into the wells of the ELISA plate.
- The ELISA plate was incubated with shaking at 37° C. for 1 h, and washed with PBST. Then, 40 ng/ml of Goat anti-Human IgG Fc Cross-Adsorbed Secondary Antibody-HRP (Art. No. 31413, Thermo) was added into the wells of the plate, and the plate was incubated with shaking at 37° C. for 1 h and washed with PBST. Then, TMB was added as HRP chromogenic substrate. The plate was incubated for 15 min, and then the reaction was terminated with 2N H2SO4. The absorbance at 450 nm was detected by a microplate reader. According to the substrate standard curve, the concentrations of intact or partially-cleaved trastuzumab in different test wells were calculated, and then the proportions of fully-cleaved trastuzumab to the initial trastuzumab were calculated, so as to evaluate the activity of different mutants.
- The multiple relationship between the activity of each mutant relative to that of the wild-type IdeE is shown in Table 1. The forty mutants obtained through screening in Example 1 all had higher activity than that of the wild-type IdeE, of which fifteen mutants were twice or more as active as the wild-type IdeE.
-
TABLE 1 Multiple relationships between the activity of the mutants relative to that of the wild-type IdeE measured by ELISA Fold of activity relative to wild- SEQ ID Mutant type NO: T8D 2.21 3 T8E 2.09 4 T8W 2.58 5 T8Y 2.30 6 A10K 1.99 7 A10R 2.20 8 T24A 2.23 9 T24G 2.09 10 T24S 2.18 11 A59I 2.31 12 A59L 2.41 13 A59V 2.68 14 E97D 2.00 15 R280H 2.30 16 R280K 2.20 17 — — — - Twelve mutants were selected from the fifteen mutants shown in Table 1 which were twice or more as active as the wild-type IdeE for the detection of their thermal stability. Detection method: The activity detection method was as follows:
- The supernatant of the wild-type or each mutant was divided into two parts, and the two parts were placed at 4° C. and 50° C. for 1 h, respectively. Subsequently, the activity of the wild-type or each mutant was detected according to the ELISA method in Example 2. The residual activity percentage (%) of the wild-type or each mutant after being placed at 50° C. for 1 h was calculated based on the activity after being placed at 50° C./the activity after being placed at 4° C., so as to compare the thermal stability of the wild-type and each mutant.
- The multiple relationships between the thermal stability of the mutants relative to that of the wild-type IdeE are shown in Table 2. Table 2 shows that the twelve mutants all had higher thermal stability than that of the wild-type, of which seven mutants had thermal stability more than 3 times that of the wild-type.
-
TABLE 2 Multiple relationships between the thermal stability of the mutants relative to that of the wild-type IdeE Fold of thermal stability relative SEQ ID Mutant to wild-type NO: T8D 4.14 3 T8W 4.43 5 T24A 4.50 9 A59L 3.83 13 A59V 5.20 14 E97D 4.27 15 R280H 4.36 16 — — — - The activity of cleaving human IgG1 of the seven single-point mutants as shown in Tables 1 and 2, TBD, TBW, T24A, A59L, A59V, E97D and R280H, was detected, “whose activity was more than twice that of the wild-type IdeE, and whose thermal stability was more than 3 times that of the wild-type IdeE”.
- 1. Expression and Purification of Mutants
- From each of the plates transformed with the above-mentioned five single-point mutants in Example 1, one single colony was picked, inoculated to 3 ml of LB medium containing 100 ug/ml of ampicillin, and cultured at 37° C. and 250 rpm overnight. The overnight cultures were inoculated to 50 ml of LB medium containing 100 ug/ml of ampicillin, and cultured at 37° C. until OD600 reached 0.4-0.6. Then, 0.1 mM of IPTG was added, and the mixtures were further cultured at 30° C. overnight. Supernatants were harvested by centrifugation from the overnight cultures. The supernatants were further purified with IDA-Ni agarose magnetic beads, and the purified and eluted proteins were buffer exchanged into PBS buffer system with an ultrafiltration centrifuge tube. SDS-PAGE was used to evaluate the purity of the purified mutant proteins. OD280 was detected, and the concentration of the purified mutant proteins was calculated according to the extinction coefficient.
- 2. Comparison of Human IgG1-Cleaving Activity of Mutants
- The activity of cleaving human IgG1 of different mutants relative to the wild-type IdeE was further evaluated via the cleavage products resulting from cleavage of human IgG1 by each mutant at different concentrations displayed on SDS-PAGE. The purified mutants and the wild-type IdeE were diluted to 0.002 mg/mL and 0.001 mg/mL, respectively. 50 ul of mutants or wild-type IdeE at different concentrations was each added to 50 ul of reaction system containing 2 mg/ml of trastuzumab to initiate the cleavage reaction, and the reaction systems were placed at 37° C. for 30 min. The samples were mixed with an equal volume of 2×SDS loading buffer, and then placed in water bath at 75° C. for 5 min. The cleavage products were detected by SDS-PAGE.
-
FIG. 1 shows the electrophoretogram of cleavage products resulting from cleavage of human IgG1 by the seven single-point mutants and the wild-type IdeE (enzyme:substrate=1:1000).FIG. 2 shows the electrophoretogram of cleavage products resulting from cleavage of human IgG1 by the seven single-point mutants and the wild-type IdeE (enzyme:substrate=1:2000). The seven single-point mutants at the concentration of 0.001 mg/ml all cleaved IgG1 with an efficiency not worse than that of 0.002 mg/ml of wild-type IdeE, indicating that the activity of cleaving human IgG1 of the seven single-point mutants is not less than twice that of the wild-type IdeE. - On the basis of the wild-type IdeE, the first 15 amino acids (D1-V15), the first 16 amino acids (D1-P16), the first 17 amino acids (D1-H17), the first 18 amino acids (D1-Q18) and the first 19 amino acids (D1419) at its N-terminus were respectively deleted, so as to construct five N-terminal truncated mutants (see Table 3).
-
TABLE 3 Sequence design of truncated mutants Mutant Modification relative to wild-type or mutant sequence SEQ ID NO: WT_del15 Deletion of the first 15 amino acids in SEQ ID NO: 2 18 WT_del16 Deletion of the first 16 amino acids in SEQ ID NO: 2 19 WT_del17 Deletion of the first 17 amino acids in SEQ ID NO: 2 20 WT_del18 Deletion of the first 18 amino acids in SEQ ID NO: 2 21 WT_del19 Deletion of the first 19 amino acids in SEQ ID NO: 2 22 - 1. Expression and Purification of Mutants
- According to the method in Example 1, the polynucleotide sequences of the mutants in Table 3 were synthesized, and the mutant expression recombinant plasmids were constructed to transform Escherichia coli BL21 Star (DE3). The mutant purified proteins were prepared according to the method in Example 4.
- 2. Comparison of Human IgG1-Cleaving Activity of Mutants
- The purified mutants and the wild-type IdeE were diluted to 0.002 mg/mL, respectively. 50 ul of diluted mutants or wild-type IdeE was each added to 50 ul of reaction system containing 2 mg/ml of trastuzumab to initiate the cleavage reaction, and the reaction systems were placed at 37° C. for 30 min. The samples were mixed with an equal volume of 2×SDS loading buffer, and then placed in water bath at 75° C. for 5 min. The cleavage products were detected by SDS-PAGE.
-
FIG. 3 shows the electrophoretogram of cleavage products resulting from cleavage of human IgG1 by the five truncated mutants (enzyme:substrate=1:1000). The cleavage activity of all of the five truncated mutants was not significantly different from that of the wild-type IdeE. - 3. Comparison of Thermal Stability of Mutants
- The purified mutants and the wild-type IdeE were respectively diluted to 0.1 mg/ml, placed at 50° C. for 1 h, and then further diluted to 0.002 mg/mL. 50 ul of diluted mutants or wild-type IdeE was each added to 50 ul of reaction system containing 2 mg/ml of trastuzumab to initiate the cleavage reaction, and the reaction systems were placed at 37° C. for 30 min. The samples were mixed with an equal volume of 2×SDS loading buffer, and then placed in water bath at 75° C. for 5 min. The cleavage products were detected by SDS-PAGE.
-
FIG. 4 shows the electrophoretogram of cleavage products resulting from cleavage of human IgG1 by the five truncated mutants and the wild-type IdeE after being placed at 50° C. for 1 h (enzyme:substrate=1:1000). The residual activity of the five truncated mutants after heat treatment at 50° C. was significantly higher than that of the wild-type, indicating that the thermal stability of all of the five truncated mutants is significantly improved compared with that of the wild-type. - On the basis of the wild-type IdeE, the last 5 amino acids (W311-S315) and the last 10 amino acids (S306-S315) at its C-terminus were respectively deleted, so as to construct two C-terminal truncated mutants (see Table 4).
-
TABLE 4 Sequence design of truncated mutants Mutant Modification relative to wild-type or mutant sequence SEQ ID NO: WT_delC5 Deletion of the last 5 amino acids in SEQ ID NO: 2 23 WT_delC10 Deletion of the last 10 amino acids in SEQ ID NO: 2 24 - 1. Expression and Purification of Mutants
- According to the method in Example 1, the polynucleotide sequences of the mutants in Table 4 were synthesized, and the mutant expression recombinant plasmids were constructed to transform Escherichia coli BL21 Star (DE3). The mutant purified proteins were prepared according to the method in Example 4.
- 2. Comparison of Human IgG1-Cleaving Activity of Mutants
- The purified mutants and the wild-type IdeE were diluted to 0.002 mg/mL, respectively. 50 ul of diluted mutants or wild-type IdeE was each added to 50 ul of reaction system containing 2 mg/ml of trastuzumab to initiate the cleavage reaction, and the reaction systems were placed at 37° C. for 30 min. The samples were mixed with an equal volume of 2×SDS loading buffer, and then placed in water bath at 75° C. for 5 min. The cleavage products were detected by SDS-PAGE.
-
FIG. 5 shows the electrophoretogram of cleavage products resulting from cleavage of human IgG1 by the two C-terminal truncated mutants (enzyme:substrate=1:1000). The cleavage activity of both of the two truncated mutants was more than twice that of the wild-type IdeE. - On the basis of the five single-point mutants, T24A, A59L, A59V, E97D and R280H, the first 18 amino acids (D1-Q18) were respectively deleted, so as to construct five combinatorial mutants (see Table 5).
-
TABLE 5 Sequence design of combinatorial mutants Mutant Modification relative to wild-type or mutant sequence SEQ ID NO: T24A_del18 Deletion of the first 18 amino acids in SEQ ID NO: 9 25 A59L_del18 Deletion of the first 18 amino acids in SEQ ID NO: 13 26 A59V_del18 Deletion of the first 18 amino acids in SEQ ID NO: 14 27 E97D_del18 Deletion of the first 18 amino acids in SEQ ID NO: 15 28 R280H_del18 Deletion of the first 18 amino acids in SEQ ID NO: 16 29 - 1. Expression and Purification of Mutants
- According to the method in Example 1, the polynucleotide sequences of the mutants in Table 5 were synthesized, and the mutant expression recombinant plasmids were constructed to transform Escherichia coli BL21 Star (DE3). The mutant purified proteins were prepared according to the method in Example 4.
- 2. Comparison of Human IgG1-Cleaving Activity of Mutants
- The purified mutants were diluted to 0.001 mg/mL, respectively. 50 ul of diluted mutants or wild-type IdeE was each added to 50 ul of reaction system containing 2 mg/ml of trastuzumab to initiate the cleavage reaction, and the reaction systems were placed at 37° C. for 30 min. The samples were mixed with an equal volume of 2×SDS loading buffer, and then placed in water bath at 75° C. for 5 min. The cleavage products were detected by SDS-PAGE.
-
FIG. 6 shows the electrophoretogram of cleavage products resulting from cleavage of human IgG1 by the five combinatorial mutants (enzyme:substrate=1:2000). Comparing the cleavage inFIG. 6 to that inFIG. 2 , there was no significant difference in cleavage activity between the five truncated mutants and the five single-point combinatorial mutants, indicating that the activity of cleaving human IgG1 of the combinatorial mutants is also not less than twice that of the wild-type IdeE. - 3. Comparison of Thermal Stability of Mutants
- The purified mutants were respectively diluted to 0.1 mg/ml, placed at 50° C. for 1 h, and then further diluted to 0.001 mg/mL. 50 ul of diluted mutants or wild-type IdeE was each added to 50 ul of reaction system containing 2 mg/ml of trastuzumab to initiate the cleavage reaction, and the reaction systems were placed at 37° C. for 30 min. The samples were mixed with an equal volume of 2×SDS loading buffer, and then placed in water bath at 75° C. for 5 min. The cleavage products were detected by SDS-PAGE.
-
FIG. 7 shows the electrophoretogram of cleavage products resulting from cleavage of human IgG1 by the five combinatorial mutants after being placed at 50° C. for 1 h (enzyme:substrate=1:2000). Comparing the cleavage inFIG. 8 to that inFIG. 7 , the activity of the five combinatorial mutants after heat treatment at 50° C. only decreased slightly, indicating that the thermal stability of all of the five combinatorial mutants is also significantly improved compared with that of the wild-type. - The purified E97D_del18 mutant in Example 7 was sequentially diluted to 20 μg/mL, 10 μg/mL, 5 μg/mL, 2.5 μg/mL and 1.25 μg/ml. IdeS (FabRICATOR®, Art. No. AO-FRI-020, Genovis) was respectively diluted to 2 U/μl, 1 U/μl, 0.5 U/μl, 0.25 U/μl and 0.125 U/μl according to its label. IdeZ (FabRICATOR-Z®, Art. No. AO-FRZ-020, Genovis) was diluted to 0.4 U/μl, 0.2 U/μl, 0.1 U/μl, 0.05 U/μl and 0.025 U/μl, respectively. 50 μl of mutants, IdeS or IdeZ at different concentrations was each added to 50 μl of reaction system containing 2 mg/ml of trastuzumab to initiate the cleavage reaction, and the reaction systems were placed at 37° C. for 30 min. The samples were mixed with an equal volume of 2×SDS loading buffer, and then placed in water bath at 75° C. for 5 min. The cleavage products were detected by SDS-PAGE.
-
FIG. 8 shows the electrophoretogram of cleavage products resulting from cleavage of human IgG1 by mutant E97D_del18 and IdeS at different concentrations. From the enzyme protein bands on the electrophoretogram, it can be judged that the concentration of enzyme IdeS inlane 1 was between those of mutant enzyme E97D_del18 inlanes lane 3 was deduced to be between those of mutant enzyme E97D_del18 inlanes lane 3 was between those inlanes -
FIG. 9 shows the electrophoretogram of cleavage products resulting from cleavage of human IgG1 by mutant E97D_del18 and IdeZ at different concentrations. From the enzyme protein bands on the electrophoretogram, it can be judged that the concentration of enzyme IdeZ inlane 1 was higher than that of mutant enzyme E97D_del18 inlane 7, thus the concentration of enzyme IdeZ inlane 3 was deduced to be higher than that of mutant enzyme E97D_del18 inlane 9, that is, 4 times higher than that of mutant enzyme E97D_del18 inlane 11, and the enzymatic digestion of IgG1 inlane 3 was close to that inlane 11, and therefore it can be inferred that the activity of cleaving human IgG1 of mutant E97D_del18 is 4 times higher than that of IdeZ. - The activity of cleaving human IgG1 in vitro of mutant E97D_del18 was evaluated by detecting the amount of intact or single-cleaved IVIg in the sera or plasmas of mice treated with mutant E97D_del18 and human IVIg. According to Table 6, mouse serum or plasma enzymatic digestion systems were formulated for different groups.
-
TABLE 6 Mouse serum or plasma enzymatic digestion systems Concentration Volume Concentration Concentration of iodoacetic Volume of serum of IVIg in of E97D_del18 acid in the of PBS or plasma the system in the system system Group name (μl) (μl) (mg/ml) (mg/ml) (mM) IVIg control 100 / 10 / / Mouse serum / 100 10 / / control group Mouse serum / 100 10 0.05 / normal enzymatic digestion group Mouse serum / 100 10 0.05 2 iodoacetic acid treatment group Mouse plasma / 100 10 / / control group Mouse plasma / 100 10 0.05 / normal enzymatic digestion group Mouse plasma / 100 10 0.05 2 iodoacetic acid treatment group - The effect of iodoacetic acid in the iodoacetic acid treatment groups was to inhibit the activity of the IgG-degrading enzyme.
- The systems were placed at 37° C. for 30 min. 20 μl of samples were mixed with an equal volume of 2×SDS non-reducing loading buffer, and then placed in water bath at 75° C. for 5 min. The cleavage products were detected by SDS-PAGE.
-
FIG. 10 shows the electrophoretogram of cleavage products resulting from cleavage of human IVIg by mutant E97D_del18 in the sera and plasmas of mice. The results showed that E97D_del18 can effectively cleave human IVIg in both the sera and plasmas of mice. - Whether mutant E97D_del18 has the activity of cleaving human IgG1 in vitro was evaluated by detecting the sera of mice or humans treated with mutant E97D_del18. According to Table 7, mouse or human serum enzymatic digestion systems were formulated for different groups.
-
TABLE 7 Mouse or human serum enzymatic digestion systems Volume of Concentration of Volume of normal human E97D_del18 mouse serum serum in the system Group name (μl) (μl) (mg/ml) Mouse serum control 100 / / group Mouse serum 100 / / enzymatic digestion group Human serum control / 100 0.02 group Human serum / 100 0.02 enzymatic digestion group - The systems were placed at 37° C. for 24 h. 20 μl of samples were mixed with an equal volume of 2×SDS reducing loading buffer, then diluted 20× with 1×SDS reducing loading buffer, and placed in water bath at 75° C. for 5 min. The cleavage products were detected by SDS-PAGE.
-
FIG. 11 shows the electrophoretogram of cleavage products produced by mutant E97D_del18 in the sera of mice and humans. The results showed that a significantly visible 25 kD Fc fragment was produced in the sera of humans through E97D_del18 cleavage, which was invisible in the sera of mice, indicating that E97D_del18 can effectively and specifically cleave IgG1 in the sera of humans, and has very low or no activity of cleaving IgG1 in the sera of mice. - The activity of cleaving serum immunoglobulins of different species of animals in vitro of mutant E97D_del18 was evaluated by detecting the amount of intact or single-cleaved IgG in the sera or plasmas of different species of animals added with mutant E97D_del18. According to Tables 8 and 9, serum or antibody enzymatic digestion systems were formulated for different species.
-
TABLE 8 Beagle serum and antibody (of different species) enzymatic digestion systems Concentration of Concentration of immunoglobulin E97D_del18 in the system in the system Group name (mg/ml) (mg/ml) Beagle serum control group 10 / Beagle serum enzymatic digestion group 10 0.025/1 Rabbit polyclonal IgG antibody 1 / 1 control group Rabbit polyclonal IgG antibody 1 0.005 1 control group Rabbit polyclonal IgG antibody 1 / 2 control group Rabbit polyclonal IgG antibody 1 0.005 2 control group Mouse monoclonal antibody IgG1 1 0.005 control group Mouse monoclonal antibody IgG1 1 0.005 enzymatic digestion group Mouse monoclonal antibody IgG2a 1 0.005 control group Mouse monoclonal antibody IgG2a 1 0.005 enzymatic digestion group - The systems were placed at 37° C. for 1 h. The enzymatic digestion products were detected by SDS-PAGE.
-
TABLE 9 Serum enzymatic digestion systems of different species Concentration of Concentration of immunoglobulin E97D_del18 in the system in the system Group name (mg/ml) (mg/ml) SD rat serum control group 5 / SD rat serum enzymatic digestion group 5 0.025/0.5 ICR mouse serum control group 5 / ICR mouse serum enzymatic digestion group 5 0.025/0.5 New Zealand rabbit serum control group 5 / New Zealand rabbit serum enzymatic 5 0.025/0.5 digestion group Beagle serum control group 5 / Beagle serum enzymatic digestion group 5 0.025/0.5 Cynomolgus monkey serum control group 5 / Cynomolgus monkey serum enzymatic 5 0.025/0.5 digestion group Bama mini-pig serum control group 5 / Bama mini-pig serum enzymatic digestion 5 0.025/0.5 group -
FIGS. 12A-12D show the cleavage activity of mutant E97D_del18 on the sera and antibodies of different species. The results showed that E97D_del18 can effectively cleave dog IgG, rabbit IgG and mouse IgG2a, but can not cleave mouse IgG1. E97D_del18 can effectively cleave IgG in the sera of rabbits, dogs and monkeys, of which IgG in the sera of rabbits was cleaved most effectively, IgG in the sera of pigs was cleaved more effectively, and IgG in the sera of rats and mice was nearly uncleaved. - The assay was based on the competition between mutant E97D_del18 and IdeS for binding to anti-E97D_del18/IdeS antibodies. Pre-incubation of test enzymes and human serum would enable anti-E97D_del18/IdeS antibodies to bind to mutant E97D_del18 and IdeS.
- A well plate was coated with mutant E97D_del18 and IdeS overnight, then washed with PBST, and blocked in 2% BSA blocking solution for 1 h. A mixed plate was prepared with the mutant to be tested and IdeS diluted stepwise and human serum. The mixed plate was incubated with shaking at room temperature for 1 h, and washed with PBST. Then, biotin-labeled E97D_del18 mutant and IdeS were added, followed by SA-HRP. The plate was developed with TMB, and read. Parallel comparison obtained the presence of pre-existing antibodies against E97D_del18 and IdeS in about eighty human blood samples.
- The results are shown in Table 10, showing that the proportion of pre-existing antibodies against IdeS in normal human serum was as high as about 90%, while for mutant E97D_del18, it was only about 20%. The pre-existing antibodies against mutant E97D_del18 in vivo were significantly less than those against IdeS, demonstrating that mutant E97D_del18 has lower immunogenicity, and is more conducive to in vivo administration.
-
TABLE 10 Comparison of pre-existing antibodies against mutant E97D_del18 and IdeS in human blood samples Mutant E97D_del18 Ides Total number of human blood 76 76 samples (cases) Proportion of samples positive 18.4 89.5 for pre-existing antibodies (%) - Under sterile conditions, two mice were injected with human IVIg (an intravenous human immunoglobulin) at a dose of 1 g/kg intraperitoneally (two mice were parallel experiments, numbered No. 1 and No. 2). 24 h after injecting human IVIg, the mice were injected with the IgG-degrading enzyme (E97D_del18) at a dose of 5 mg/kg intravenously. 0 h, 15 min, 2 h, 6 h and 24 h after injecting E97D_del18, blood was collected from the two mice, and serum samples were collected, respectively. 20 μl of serum samples were mixed with an equal volume of 2×SDS non-reducing loading buffer, then diluted 20× with 1×SDS non-reducing loading buffer, and placed in water bath at 75° C. for 5 min. The cleavage products were detected by SDS-PAGE.
-
FIG. 13 shows the electrophoretogram of cleavage products resulting from cleavage of human IVIg by E97D_del18 at different times in the mice. The results showed that E97D_del18 effectively cleaved IVIg in the mice, and the enzymatic digestion was already substantially complete in 15 min. - On the basis of the above-mentioned mutants, six combinatorial mutants were further constructed, the sequences of which are shown in Table 11.
-
TABLE 11 Combinatorial mutants Modification relative to wild-type or mutant Mutant sequence SEQ ID NO: E97D_del18_delC5 Deletion of the last 5 amino acids in SEQ ID NO: 28 30 E97D_del18_delC10 Deletion of the last 10 amino acids in SEQ ID NO: 28 31 A59V_del18_delC5 Deletion of the last 5 amino acids in SEQ ID NO: 27 32 A59L_del18_delC5 Deletion of the last 5 amino acids in SEQ ID NO: 26 33 R280H_del18_delC5 Deletion of the last 5 amino acids in SEQ ID NO: 29 34 E97D_A59V_R280H Combination of three mutations, E97D, A59V and 35 R280H - 1. Expression and Purification of Mutants
- According to the method in Example 1, the polynucleotide sequences of the mutants in Table 11 were synthesized, and the mutant expression recombinant plasmids were constructed to transform Escherichia coli BL21 Star (DE3). The mutant purified proteins were prepared according to the method in Example 4.
- 2. Comparison of Human IgG1-Cleaving Activity of Mutants
- The purified mutants were diluted to 0.001 mg/mL, respectively. 50 ul of diluted mutants or wild-type IdeE was each added to 50 ul of reaction system containing 2 mg/ml of trastuzumab to initiate the cleavage reaction, and the reaction systems were placed at 37° C. for 30 min. The samples were mixed with an equal volume of 2×SDS loading buffer, and then placed in water bath at 75° C. for 5 min. The cleavage products were detected by SDS-PAGE.
-
FIGS. 14A and 14B show the electrophoretograms of cleavage products resulting from cleavage of human IgG1 by the six combinatorial mutants (enzyme:substrate=1:2000). - The Applicant declares that the detailed methods of the present invention are illustrated by means of the above-mentioned examples, but the present invention is not limited to the detailed methods mentioned above, that is, it does not mean that the present invention must rely on the detailed methods mentioned above to carry out. Those skilled in the art should understand that any improvement of the present invention, the equivalent replacement of each raw material of the articles of manufacture of the present invention, the addition of auxiliary components, the selection of specific methods, etc., fall within the scope of protection and the scope of disclosure of the present invention.
Claims (18)
1. A mutant of an immunoglobulin-degrading enzyme IdeE, wherein the immunoglobulin-degrading enzyme IdeE comprises or consists of an amino acid sequence as set forth in SEQ ID NO: 2 in the Sequence Listing; and the mutant comprises a mutation selected from the group consisting of:
(1) substitution of one or more of positions 8, 10, 24, 59, 97 and 280 of the amino acid sequence, thereby obtaining the mutant; and/or
(2) truncation of the immunoglobulin-degrading enzyme IdeE, by deleting the sequence of the first 1, the first 2, the first 3, the first 4, the first 5, the first 6, the first 7, the first 8, the first 9, the first 10, the first 11, the first 12, the first 13, the first 14, the first 15, the first 16, the first 17, the first 18 or the first 19 amino acids at its N-terminus; and/or
(3) truncation of the immunoglobulin-degrading enzyme IdeE, by deleting the sequence of the last 1, the last 2, the last 3, the last 4, the last 5, the last 6, the last 7, the last 8, the last 9 or the last 10 amino acids at its C-terminus;
wherein the mutant has higher activity than that of the immunoglobulin-degrading enzyme IdeE, and/or has higher thermal stability than that of the immunoglobulin-degrading enzyme IdeE.
2. The mutant according to claim 1 , wherein the mutation is selected from the group consisting of:
(1) substitution of position 8, 10, 24, 59, 97 or 280 of the amino acid sequence as set forth in SEQ ID NO: 2; and/or
(2) deletion of the first 15, the first 16, the first 17, the first 18 or the first 19 amino acids at the N-terminus of the immunoglobulin-degrading enzyme IdeE, preferably the first 18 amino acids; and/or
(3) deletion of the last 5 or the last 10 amino acids at the C-terminus of the immunoglobulin-degrading enzyme IdeE, preferably the last 5 amino acids.
3. The mutant according to claim 1 , wherein the substitution is selected from the group consisting of:
(1) the threonine at position 8 is substituted with any one of cysteine, phenylalanine, tryptophan, tyrosine, aspartic acid, glutamic acid, alanine, glycine, histidine, isoleucine, leucine, methionine, asparagine, proline, glutamine, serine, valine, arginine and lysine;
(2) the alanine at position 10 is substituted with any one of cysteine, aspartic acid, glutamic acid, phenylalanine, glycine, histidine, isoleucine, lysine, leucine, methionine, asparagine, proline, glutamine, arginine, serine, threonine, valine, tryptophan and tyrosine;
(3) the threonine at position 24 is substituted with any one of alanine, cysteine, aspartic acid, asparagine, glutamic acid, phenylalanine, glycine, histidine, isoleucine, lysine, leucine, methionine, proline, glutamine, arginine, serine, valine, tryptophan and tyrosine;
(4) the alanine at position 59 is substituted with any one of cysteine, aspartic acid, glutamic acid, phenylalanine, glycine, histidine, isoleucine, lysine, leucine, methionine, asparagine, proline, glutamine, arginine, serine, threonine, valine, tryptophan and tyrosine;
(5) the glutamic acid at position 97 is substituted with any one of alanine, cysteine, aspartic acid, phenylalanine, glycine, histidine, isoleucine, lysine, leucine, methionine, asparagine, proline, glutamine, arginine, serine, threonine, valine, tryptophan and tyrosine; and
(6) the arginine at position 280 is substituted with any one of alanine, aspartic acid, glutamic acid, cysteine, serine, phenylalanine, histidine, isoleucine, lysine, leucine, methionine, asparagine, proline, glutamine, tryptophan, threonine, valine and tyrosine.
4. The mutant according to claim 3 , wherein the mutant is as set forth in any one of SEQ ID NOs: 3-35 in the Sequence Listing.
5. (canceled)
6. (canceled)
7. A nucleotide encoding the mutant according to claim 1 .
8. An expression vector, comprising the nucleotide according to claim 7 .
9. A host cell, comprising the expression vector according to claim 8 , preferably an E. coli cell or a yeast cell.
10. A composition, comprising:
an immunoglobulin-degrading enzyme or a mutant thereof or a protein comprising the immunoglobulin-degrading enzyme or the mutant thereof; and
optionally, a pharmaceutically acceptable carrier or excipient.
11. The composition according to claim 10 , wherein the immunoglobulin-degrading enzyme is selected from IdeE, IdeS and IdeZ.
12. The composition according to claim 10 , wherein the mutant of the immunoglobulin-degrading enzyme is a mutant of an immunoglobulin-degrading enzyme IdeE, wherein the immunoglobulin-degrading enzyme IdeE comprises or consists of an amino acid sequence as set forth in SEQ ID NO: 2 in the Sequence Listing; and the mutant comprises a mutation selected from the group consisting of:
(1) substitution of one or more of positions 8, 10, 24, 59, 97 and 280 of the amino acid sequence, thereby obtaining the mutant; and/or
(2) truncation of the immunoglobulin-degrading enzyme IdeE, by deleting the sequence of the first 1, the first 2, the first 3, the first 4, the first 5, the first 6, the first 7, the first 8, the first 9, the first 10, the first 11, the first 12, the first 13, the first 14, the first 15, the first 16, the first 17, the first 18 or the first 19 amino acids at its N-terminus; and/or
(3) truncation of the immunoglobulin-degrading enzyme IdeE, by deleting the sequence of the last 1, the last 2, the last 3, the last 4, the last 5, the last 6, the last 7, the last 8, the last 9 or the last 10 amino acids at its C-terminus;
wherein the mutant has higher activity than that of the immunoglobulin-degrading enzyme IdeE, and/or has higher thermal stability than that of the immunoglobulin-degrading enzyme IdeE.
13. The composition according to claim 10 , further comprising:
an antibody or an Fc-containing protein.
14. The composition according to claim 13 , wherein the antibody target is selected from the group consisting of: a cell surface protein, a cytokine, a hormone, an enzyme, an intracellular messenger, an intercellular messenger and an immune checkpoint.
15. The composition according to claim 10 , further comprising:
a viral vector drug, preferably selected from the group consisting of: an oncolytic virus, a gene therapy virus and a viral vector vaccine.
16. The composition according to claim 10 , further comprising:
an agent capable of reducing the IgG level in the blood, preferably selected from the group consisting of: an FcRn antibody and an Fc fragment variant with a high affinity to FcRn.
17. A kit, comprising:
(1) the mutant according to claim 1 ; and
(2) one or more selected from the group consisting of: (a) a pharmaceutically acceptable carrier or excipient; and (b) an antibody or an Fc-containing protein; and/or
(3) a viral vector drug selected from an oncolytic virus, a gene therapy virus and a viral vector vaccine; and/or
(4) an agent capable of reducing the IgG level in the blood selected from an FcRn antibody and an Fc fragment variant with a high affinity to FcRn.
18. A kit, comprising: a part A and a part B, wherein
the part A comprises the mutant according to claim 1 , and
the part B comprises one or more selected from the group consisting of:
(1) a pharmaceutically acceptable carrier or excipient; (2) an antibody or an Fc-containing protein; and/or (3) a viral vector drug; and/or (4) an agent capable of reducing the IgG level in the blood;
wherein the viral vector drug is selected from an oncolytic virus, a gene therapy virus and a viral vector vaccine; and the agent capable of reducing the IgG level in the blood is selected from an FcRn antibody and an Fc fragment variant with a high affinity to FcRn.
Applications Claiming Priority (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202010557830.X | 2020-06-18 | ||
CN202010557830 | 2020-06-18 | ||
PCT/CN2021/100844 WO2021254479A1 (en) | 2020-06-18 | 2021-06-18 | Mutant of immunoglobulin degrading enzyme idee |
Publications (1)
Publication Number | Publication Date |
---|---|
US20230364207A1 true US20230364207A1 (en) | 2023-11-16 |
Family
ID=79268498
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US18/001,876 Pending US20230364207A1 (en) | 2020-06-18 | 2021-06-18 | Mutant of immunoglobulin degrading enzyme idee |
Country Status (5)
Country | Link |
---|---|
US (1) | US20230364207A1 (en) |
EP (1) | EP4169934A4 (en) |
JP (1) | JP2023532219A (en) |
CN (2) | CN115443288A (en) |
WO (1) | WO2021254479A1 (en) |
Families Citing this family (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN118382456A (en) | 2021-12-16 | 2024-07-23 | 上海宝济药业股份有限公司 | Fc variants resistant to cleavage by immunoglobulin degradation enzymes |
TW202334183A (en) * | 2021-12-22 | 2023-09-01 | 大陸商上海寶濟藥業有限公司 | Use of mutant of immunoglobulin degrading enzyme idee |
CN114875003B (en) * | 2022-04-06 | 2024-05-24 | 浙江大学 | Mutant of short-chain dehydrogenase, coding gene obtaining method and application of mutant |
EP4349365A1 (en) | 2022-10-07 | 2024-04-10 | Hansa Biopharma AB | Co-treatment for gene therapy |
CN118497176A (en) * | 2024-07-16 | 2024-08-16 | 苏州康聚生物科技有限公司 | Immunoglobulin degrading enzyme |
Family Cites Families (9)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP2227249B1 (en) * | 2007-12-13 | 2018-04-25 | Intervacc AB | Improved immunizing composition |
EP3135684B1 (en) * | 2010-05-26 | 2023-06-28 | Intervacc AB | Vaccine against streptococcal infections based on recombinant fusion proteins |
GB201502305D0 (en) * | 2015-02-12 | 2015-04-01 | Hansa Medical Ab | Protein |
GB201502306D0 (en) * | 2015-02-12 | 2015-04-01 | Hansa Medical Ab | Protein |
US11155585B2 (en) * | 2015-07-09 | 2021-10-26 | Intervacc Ab | Vaccine against S. suis infection |
KR102546854B1 (en) * | 2015-07-16 | 2023-06-22 | 다이이찌 산쿄 가부시키가이샤 | Novel endos mutant enzyme |
SG11201806639VA (en) * | 2016-02-04 | 2018-09-27 | Genovis Ab | New streptococcal proteases |
WO2018093868A1 (en) * | 2016-11-16 | 2018-05-24 | University Of Florida Research Foundation, Inc. | Immunoglobulin proteases, compositions, and uses thereof |
US20230374083A1 (en) * | 2020-09-21 | 2023-11-23 | Shanghai Bao Pharmaceuticals Co., Ltd. | Pharmaceutical combination and use thereof |
-
2021
- 2021-06-18 CN CN202180013436.2A patent/CN115443288A/en active Pending
- 2021-06-18 WO PCT/CN2021/100844 patent/WO2021254479A1/en unknown
- 2021-06-18 CN CN202410654527.XA patent/CN118562771A/en active Pending
- 2021-06-18 US US18/001,876 patent/US20230364207A1/en active Pending
- 2021-06-18 JP JP2022577764A patent/JP2023532219A/en active Pending
- 2021-06-18 EP EP21825273.2A patent/EP4169934A4/en active Pending
Also Published As
Publication number | Publication date |
---|---|
CN118562771A (en) | 2024-08-30 |
CN115443288A (en) | 2022-12-06 |
WO2021254479A1 (en) | 2021-12-23 |
EP4169934A4 (en) | 2024-07-24 |
EP4169934A1 (en) | 2023-04-26 |
JP2023532219A (en) | 2023-07-27 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US20230364207A1 (en) | Mutant of immunoglobulin degrading enzyme idee | |
EP3854806A1 (en) | Novel interleukin 2 and use thereof | |
CA3055202A1 (en) | Cd19 compositions and methods for immunotherapy | |
JP2019088290A (en) | ASGPR antibodies and uses thereof | |
JP2019033755A (en) | Interleukin-10 fusion proteins and uses thereof | |
CA3038526A1 (en) | Immunomodulatory fusion proteins | |
JP7308034B2 (en) | Optimized double nuclease fusions and methods | |
AU2016253126A1 (en) | Kappa myeloma antigen chimeric antigen receptors and uses thereof | |
JP2017506075A (en) | Interleukin-10 immunoconjugate | |
AU2021225156B2 (en) | UTI fusion proteins | |
US20240336908A1 (en) | Use of mutant of immunoglobulin degrading enzyme idee | |
KR102647165B1 (en) | A universal self-regulating mammalian cell line platform for biologics production | |
WO2021146487A2 (en) | Il2 orthologs and methods of use | |
KR20240046224A (en) | Bispecific antibodies and their uses | |
CA3234552A1 (en) | Heterodimeric fc cytokines and uses thereof | |
CA3100112A1 (en) | Engineering b lymphocytes by utilizing endogenous activation-induced cytidine deaminase | |
EP4419545A2 (en) | Heterodimeric fc cytokines and uses thereof | |
CN113769058B (en) | Pharmaceutical composition and application thereof | |
WO2023138643A1 (en) | Cd39 antigen binding protein and use thereof | |
CN118359730A (en) | Targeted fusion proteins comprising cytokines | |
CN118530362A (en) | Herceptin antibody mutant and NK cell coupled with herceptin antibody mutant | |
KR20240156649A (en) | Systems and methods to produce b cells genetically modified to express selected antibodies |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
AS | Assignment |
Owner name: SHANGHAI BAO PHARMACEUTICALS CO., LTD., CHINA Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LIU, YANJUN;WANG, ZHENG;REEL/FRAME:062983/0139 Effective date: 20230118 |
|
STPP | Information on status: patent application and granting procedure in general |
Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION |